Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA...

23
Multifunctional DNA nanomaterials for biomedical applications Tam, Dick Yan; Lo, Pik Kwan Published in: Journal of Nanomaterials Published: 01/01/2015 Document Version: Final Published version, also known as Publisher’s PDF, Publisher’s Final version or Version of Record License: CC BY Publication record in CityU Scholars: Go to record Published version (DOI): 10.1155/2015/765492 Publication details: Tam, D. Y., & Lo, P. K. (2015). Multifunctional DNA nanomaterials for biomedical applications. Journal of Nanomaterials, 2015, [765492]. https://doi.org/10.1155/2015/765492 Citing this paper Please note that where the full-text provided on CityU Scholars is the Post-print version (also known as Accepted Author Manuscript, Peer-reviewed or Author Final version), it may differ from the Final Published version. When citing, ensure that you check and use the publisher's definitive version for pagination and other details. General rights Copyright for the publications made accessible via the CityU Scholars portal is retained by the author(s) and/or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Users may not further distribute the material or use it for any profit-making activity or commercial gain. Publisher permission Permission for previously published items are in accordance with publisher's copyright policies sourced from the SHERPA RoMEO database. Links to full text versions (either Published or Post-print) are only available if corresponding publishers allow open access. Take down policy Contact [email protected] if you believe that this document breaches copyright and provide us with details. We will remove access to the work immediately and investigate your claim. Download date: 21/08/2020

Transcript of Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA...

Page 1: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Multifunctional DNA nanomaterials for biomedical applications

Tam Dick Yan Lo Pik Kwan

Published inJournal of Nanomaterials

Published 01012015

Document VersionFinal Published version also known as Publisherrsquos PDF Publisherrsquos Final version or Version of Record

LicenseCC BY

Publication record in CityU ScholarsGo to record

Published version (DOI)1011552015765492

Publication detailsTam D Y amp Lo P K (2015) Multifunctional DNA nanomaterials for biomedical applications Journal ofNanomaterials 2015 [765492] httpsdoiorg1011552015765492

Citing this paperPlease note that where the full-text provided on CityU Scholars is the Post-print version (also known as Accepted AuthorManuscript Peer-reviewed or Author Final version) it may differ from the Final Published version When citing ensure thatyou check and use the publishers definitive version for pagination and other details

General rightsCopyright for the publications made accessible via the CityU Scholars portal is retained by the author(s) andor othercopyright owners and it is a condition of accessing these publications that users recognise and abide by the legalrequirements associated with these rights Users may not further distribute the material or use it for any profit-making activityor commercial gainPublisher permissionPermission for previously published items are in accordance with publishers copyright policies sourced from the SHERPARoMEO database Links to full text versions (either Published or Post-print) are only available if corresponding publishersallow open access

Take down policyContact lbscholarscityueduhk if you believe that this document breaches copyright and provide us with details We willremove access to the work immediately and investigate your claim

Download date 21082020

Review ArticleMultifunctional DNA Nanomaterials forBiomedical Applications

Dick Yan Tam12 and Pik Kwan Lo12

1Department of Biology and Chemistry City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong2Shenzhen Key Laboratory of Biochip Research City University of Hong Kong Shenzhen 518057 China

Correspondence should be addressed to Pik Kwan Lo peggylocityueduhk

Received 4 July 2014 Accepted 26 August 2014

Academic Editor Daniela Predoi

Copyright copy 2015 D Y Tam and P K Lo This is an open access article distributed under the Creative Commons AttributionLicense which permits unrestricted use distribution and reproduction in any medium provided the original work is properlycited

The rapidly emerging DNA nanotechnology began with pioneer Seemanrsquos hypothesis that DNA not only can carry geneticinformation but also can be used as molecular organizer to create well-designed and controllable nanomaterials for applications inmaterials science nanotechnology and biology DNA-based self-assembly represents a versatile system for nanoscale constructiondue to the well-characterized conformation of DNA and its predictability in the formation of base pairs The structural features ofnucleic acids form the basis of constructing a wide variety of DNAnanoarchitectures with well-defined shapes and sizes in additionto controllable permeability and flexibility More importantly self-assembled DNA nanostructures can be easily functionalized toconstruct artificial functional systems with nanometer scale precision for multipurposes Apparently scientists envision artificialDNA-based nanostructures as tool for drug loading and in vivo targeted delivery because of their abilities in selective encapsulationand stimuli-triggered release of cargo Herein we summarize the strategies of creating multidimensional self-assembled DNAnanoarchitectures and review studies investigating their stability toxicity delivery efficiency loading and control release of cargosin addition to their site-specific targeting and delivery of drug or cargo molecules to cellular systems

1 Introduction

Public healthcare is a big issue among the society and hasdrawn much attention to general public In general someorganic small-molecules proteins and nucleic acids haveexhibited their promise as therapeutic agents for biomedicaltherapy In the past years scientists dreamed of improving thedelivery efficacy of these target drugs for various biologicaland biomedical applications However problems in termsof solubility toxicity cost and penetration ability need tobe solved They face several transport barriers after theyare introduced to human body before going to their sitesof action For example first drug molecules have to bestable in the circulation system passing through the bloodvessel and being recognized by those particular diseased cellsAfterwards they have to pass through the highly chargeableplasma membrane andor the nuclear membrane They alsohave to withstand the acidic cellular environment Finallythe multiple drug resistance mechanism also needs to beconsidered Thus it is of great importance developing smart

systemwhich exhibits specific targeting and has high deliveryefficacy of active drug molecules

Scientists envision the rapid development of materialsciences offering great advantage for creating smart drugdelivery vehicles or carriers Various drug delivery systemsbased on different materials have been developed [1] Forexample drugs can be loaded onto the nanoparticles [2] ornanodiamonds [3] for targeted delivery Active biomoleculardrugs can be coordinated to metals inside the carbon nan-otube and then released by heating up the nanotubes samples[4] Another advanced development is to deliver siRNA byPEGylated cyclodextrin molecules [5] They were released bydissociation of the complexes in lysosome Particularly themost commonly used drug delivery system is the polymericmaterials [6]The biblock copolymers tend to formmicelle inthe presence of drug molecules Therefore drug can be easilyloaded into the core of micelle [7] However being usefuldrug nanocarriers it is necessary to consider their toxicitybiocompatibility and stability in a cellular environment Itis well-known that most of the nanoparticles are toxic they

Hindawi Publishing CorporationJournal of NanomaterialsVolume 2015 Article ID 765492 21 pageshttpdxdoiorg1011552015765492

2 Journal of Nanomaterials

may induce cytotoxicity in living systems Heat triggered-release of drug molecules in a cellular environment is notappreciated because other healthy cells may also be affectedIn addition the efficiency and selectivity of drug loadingin polymeric micelles is also highly limited Therefore todesign new materials as drug carriers these carriers shouldhave a capability of drug incorporation and controlled releasein a highly effective way They should also be highly stableand biocompatible in a specific cellular environment It isalso necessary for them to target particular areas and carrymultifunction in order to enhance the delivery efficiency

Indeed developing novel biocompatible and multifunc-tional nanocarriers remains a key challenge for targeted drugdelivery The rapidly emerging DNA nanotechnology beganwith pioneer Seemanrsquos hypothesis that DNA not only cancarry genetic information but also can be used as molecularorganizer to create well-designed and controllable nanoma-terials for applications in materials science nanotechnologyand biology [8 9] AsDNAhas a simple and robustmolecularrecognition rule of adenine to thymine (A-T) and guanine tocytosine (G-C) pairings two complementary single-strandedDNA hybridize to form a double helix with predictableand programmable interactions The structural features ofnucleic acids form the basis of constructing a wide varietyof well-ordered DNA nanoarchitectures with well-definedshapes and sizes in addition to controllable permeabilityand flexibility [10 11] This DNA nanotechnology offersnew opportunities for the construction of complex DNAstructures in different dimensions More importantly self-assembled DNA nanostructures can be easily functionalizedto construct artificial functional systems for multipurposesApparently scientists envision artificial DNA-based nanos-tructures as tools for drug loading and in vivo targeteddelivery because of their potential of selective encapsulationand stimuli-triggered release of cargo

In this review article we concentrate on a new-comerof drug delivery carriers based on self-assembled DNAnanostructures We will demonstrate the power and promiseof DNA as a scaffold to create DNA nanostructures withprecise geometry and versatile functionality Their structuralstability in physiological conditions and internalization willbe briefly described Different cargo loadingmechanisms andtheir control release via external stimuli will be summarizedin detail As a new-comer in drug delivery system studiesof intracellular behaviorsfunctions of drug loaded DNAnanocarriers and their interactions in specific intracellularcompartments in vitro or in vivo will also be discussedSome concluding remarks will try to ascertain what the nextchallenges and outlook of this exciting research area could be

2 DNA NanotechnologyTo begin with we first briefly introduce the history and themost updated status of DNAnanotechnologyThe innovationof the field of DNA nanotechnology was first demonstratedby Seeman in the early 1980s [12] Taking advantage ofself-recognition property of DNA his group designed andconstructed modified Holliday junctions to convert one-dimensional DNA strands into branched DNA tiles with

sticky ends at the edges (Figure 1(a)) These short single-stranded units provide toeholds for further assembly of 2D-structures [13] Since then the structural role of DNA iswidely well-recognized and extensively explored Howeverthese assembly approaches did not offer rigid junctions withwell-defined angles and geometry of the final structures Toovercome these drawbacks researchers started to developadvanced rigid junctions including multicrossover [14ndash16]cross-shaped tile with arms [17] DNA tensegrity triangle[18] and parallelogram DNA tile (Figure 1(b)) [19] Withsuch unprecedented talent to construct DNA-based architec-tures highly ordered 2D-DNA surfaces with programmablearrangement and a large variety of three-dimensional poly-hedral structures were successfully assembled via sticky-end cohesion among those building blocks [20ndash22] Never-theless these tile-based assemblies have certain limitationsFor example it is difficult to control the size of resultingstructures An exact stoichiometric and a high purity controlof individual DNA fragments are still problematic for theassembly of large and complex nanostructures

Another creation in DNA nanotechnology was madeby Rothemund in 2006 [23] He invented scaffolded DNAorigami which successfully offered high complexity andversatility in DNA assembly In DNA origami a long pieceof single-stranded DNA from theM13 circular bacteriophagegenome is folded with itself into a desired pattern withthe assistance of short staple strands (Figure 1(c)) [24 25]Typical examples consist of nonperiodic 2D-structures suchas a map of the Americas stars smiley faces and otherdeliberately well-designed patterns [26 27] In this approachthe relative stoichiometric ratio on different staple strandsto a single DNA scaffold is not highly restricted Moreimportantly DNA origami is a versatile and simple one-potassembly to generate nanostructures with complex shapesof predefined dimensions as compared to the conventionalcrossover approach [28 29] In an advanced developmentKostiainenrsquos group has recently demonstrated the opticalcontrol of the DNA origami formation and release [30]Although DNA was used as the only component to guide theDNA assembly in tile-based assembly or DNA origami thisresulted in fully double-stranded and DNA-dense structures

An alternative approach to building DNA nanostructureis to bring together the programmability of DNA with func-tional and structural diversities offered by supramolecularchemistry [31]This new emerging area inDNAnanotechnol-ogy involves the insertion of synthetic molecules into DNAstrand to alter its hybridization and control the assemblyoutcome (Figure 1(d)) By conjugating synthetic moleculesat the insertion points of a DNA strand typical linearDNA duplexes can be oriented and hybridized relative toone another in a controlled manner This supramolecularDNA assembly combines the diverse structural features ofmolecules and their functionalities such as luminescenceredox magnetic and catalytic properties to generate discretewell-defined structures

Taking advantages of synthetic molecules as rigid junc-tions this can reduce the amount of DNA strands neededfor the structural definition as compared to the previoustwo methods For example Sleimanrsquos group have successfully

Journal of Nanomaterials 3

Holliday junction

H

998400H

998400H

998400HV

998400V

998400V998400V

H

H

V V

(a)TCTGATGT

ACTACA

GAGCAGCCCGTCGG

TGTACGGACATGCC

CCGTACA

GGCATGT CCGTACA

GGCATGT

TCTGATGT

ACTACA

GGCTGC

CCGACGAG

GGCTGC

CCGACGAG

ACATCATGTAGTCT

(b)

(c)

Syntheticmolecule

(d)

Figure 1 Examples of self-assembled DNA nanostructures (a) A lattice is formed by hybridization of the sticky ends of a Holliday junction(b) multistranded junction structures and crossover structures including double-crossover structure cross-shaped tile with four arms DNAtensegrity triangle and parallelogram DNA tile (c) the principle of DNA origami and the design of 2D origami formed smiling face and star(d) sequential self-assembly of hexagonal-shaped DNA nanostructure via supramolecular DNA assembly

developed DNA-conjugated m-terphenyl-based organic ver-tices for modular construction of cyclic polygons a library ofDNA polyhedral structures and nanotubes with good controlover their geometry [32] dimension [33] and flexibility [34]Besides the organic insertions other important self-assemblystrategies take advantages of transition metal- ligand- lipid-and block copolymer-based environments [35ndash37]

3 Stability of Self-AssembledDNA Nanostructures

Among various DNA assemblies three-dimensional DNAnanostructures hold promise to be the universal nanocarriers

for smart and targeted drug delivery In contrast to 1D or2D DNA structures the power of self-assembled 3D DNAnanostructures lies in their excellent stability and biocom-patibility high drug loading capability and passive deliveryinto live cells They also possess fine control over geometryprecise and monodisperse dimensions positioning of guestmolecules stimuli-responsive switching of structure andtriggered-release of cargos Typical examples of drug deliverysystems based on 3D DNA nanostructures [38 39] includetetrahedron icosahedron hexagonal barrel nanotube DNAorigami box [40] nanorobot and nanocage

To be employable as a drug carrier system in mammalsDNA nanostructures must meet several important criteria

4 Journal of Nanomaterials

Table 1 Stability of different DNA nanostructures

Linear dsDNA CpGbearingDNA tetrahedral nanostructure

A 3D multilayer rectangularparallelepiped structure

CG

CG

CG

CG

Description of the structure Normal linear DNA strand withDdeI restriction site

It is made up of four 55-merstrands extended with the CpG

sequence and a 7-meroligothymine spacer

A 3D multilayer rectangularparallelepiped structure (8 helixtimes 8 helix square lattice with

dimensions of 16 nm times 16 nm times30 nm)

Incubation temperature 37∘C 25∘C

Medium 10 FBS 50 non-heat-inactivated fetalbovine serum (FBS) Cell lysate

Decay time Decay after 08 h Start decaying after 4 h but stillnot completely decayed after 24 h Still remains stable after 12 h

Citation [41] [43] [44]

(1) they have to be stable and intact in both extracellular andintracellular environments particularly stable long enoughin the cytoplasm of cells to perform their predefined tasks(2) they should not have toxic effect in mammals and (3)the cellular immune system in mammals should toleratethe nanometer-scale DNA nanocarrier systems Thus farseveral research groups have put efforts on the stabilitystudies of DNA constructs Bermudezrsquos group indicatedthat oligonucleotide-based tetrahedral made from branchjunctions exhibit a strong resistance to enzymatic digestioncompared to the linear counterparts in terms of their decaytime constants (Table 1) [41] The reason behind this wouldhighly be due to the steric hindrance effect Since the endonu-cleases initially bind to the DNA nonspecifically with a lowaffinity and then follow by diffusion along the strands Thesteric hindrance introduced by three-dimensional tetrahe-dron would reduce the effective binding of enzymes to DNAand then inhibit DNA cleavage no matter if the enzyme actsspecifically or nonspecifically Furthermore shorter sequenceor smaller size of DNA complex can enhance the resistancetowards various nucleases as they are more difficult to bendand possibly have higher steric hindrance for the action ofthe enzymes Walsh and coworkers have demonstrated thefirst example of 3D DNA nanostructure which can enter livemammalian cells effectively with or without the help of atransfection reagent [42] They stay intact for up to 48 h incytoplasm In a recent study by Li et al they have modifiedthe tetrahedral with CpG oligonucleotides which have beenconfirmed to be taken up by macrophage RAW2647 cellseffectively (Table 1) [43]

Regarding scaffold DNA origami Mei and coworkersdemonstrated that different shapes of DNA origami nanos-tructures are stable and remain intact for 12 h after exposing

to cell lysates of various cell lines and can be easily puri-fied from lysate mixtures in contrast to single-strandedor duplex DNA (Table 1) [44] They are not accessible tovarious DNAzymes due to negatively charged large andrigid origami structures Their superior structural integrityand versatile functionality are highly preserved in relationto conventional oligonucleotides validating their use forvarious biological applications Subsequently a further studycarried out by Dietzrsquo group tested the enzymatic digestion ofDNAorigami structures [45]They are fully exposed to a largevariety of endonucleases including DNase T7 exonucleaseT7 endonuclease Msel restriction endonuclease Lambdaexonuclease and Escherichia coli exonuclease These resultsindicated that they are highly stable at 37∘C towards degra-dation as compared to duplex plasmid oligonucleotidesMore recently Schuller and his coworkers reported that CpGoligonucleotides-decorated DNA origami tubes amplify astrong immune response which are completely dependent onTLR9 stimulation in mammalian spleen cell [46]

To further optimize DNA structures in regard to enzy-matic digestion resistance Sleimanrsquos group has modified 3DDNA nanostructures using a number of chemical strategiesThey found that simple chemical modification to both endsof DNA oligos with hexanediol and hexaethylene glycol inself-assembledDNAprismatic cage or site-specific hybridiza-tion of DNA-block copolymer chains to 3D DNA scaffoldwould dramatically enhance its nuclease resistance underfetal bovine serum condition (Table 2) [47] These studiescould provide guidelines for decoration of DNA nanostruc-tures with simple chemistry modification and allow impart-ing momentous stabilization towards nuclease degradationMeanwhile the same group also demonstrated that creationof DNA nanotubes with a template generated by rolling circle

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 2: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Review ArticleMultifunctional DNA Nanomaterials forBiomedical Applications

Dick Yan Tam12 and Pik Kwan Lo12

1Department of Biology and Chemistry City University of Hong Kong Tat Chee Avenue Kowloon Hong Kong2Shenzhen Key Laboratory of Biochip Research City University of Hong Kong Shenzhen 518057 China

Correspondence should be addressed to Pik Kwan Lo peggylocityueduhk

Received 4 July 2014 Accepted 26 August 2014

Academic Editor Daniela Predoi

Copyright copy 2015 D Y Tam and P K Lo This is an open access article distributed under the Creative Commons AttributionLicense which permits unrestricted use distribution and reproduction in any medium provided the original work is properlycited

The rapidly emerging DNA nanotechnology began with pioneer Seemanrsquos hypothesis that DNA not only can carry geneticinformation but also can be used as molecular organizer to create well-designed and controllable nanomaterials for applications inmaterials science nanotechnology and biology DNA-based self-assembly represents a versatile system for nanoscale constructiondue to the well-characterized conformation of DNA and its predictability in the formation of base pairs The structural features ofnucleic acids form the basis of constructing a wide variety of DNAnanoarchitectures with well-defined shapes and sizes in additionto controllable permeability and flexibility More importantly self-assembled DNA nanostructures can be easily functionalized toconstruct artificial functional systems with nanometer scale precision for multipurposes Apparently scientists envision artificialDNA-based nanostructures as tool for drug loading and in vivo targeted delivery because of their abilities in selective encapsulationand stimuli-triggered release of cargo Herein we summarize the strategies of creating multidimensional self-assembled DNAnanoarchitectures and review studies investigating their stability toxicity delivery efficiency loading and control release of cargosin addition to their site-specific targeting and delivery of drug or cargo molecules to cellular systems

1 Introduction

Public healthcare is a big issue among the society and hasdrawn much attention to general public In general someorganic small-molecules proteins and nucleic acids haveexhibited their promise as therapeutic agents for biomedicaltherapy In the past years scientists dreamed of improving thedelivery efficacy of these target drugs for various biologicaland biomedical applications However problems in termsof solubility toxicity cost and penetration ability need tobe solved They face several transport barriers after theyare introduced to human body before going to their sitesof action For example first drug molecules have to bestable in the circulation system passing through the bloodvessel and being recognized by those particular diseased cellsAfterwards they have to pass through the highly chargeableplasma membrane andor the nuclear membrane They alsohave to withstand the acidic cellular environment Finallythe multiple drug resistance mechanism also needs to beconsidered Thus it is of great importance developing smart

systemwhich exhibits specific targeting and has high deliveryefficacy of active drug molecules

Scientists envision the rapid development of materialsciences offering great advantage for creating smart drugdelivery vehicles or carriers Various drug delivery systemsbased on different materials have been developed [1] Forexample drugs can be loaded onto the nanoparticles [2] ornanodiamonds [3] for targeted delivery Active biomoleculardrugs can be coordinated to metals inside the carbon nan-otube and then released by heating up the nanotubes samples[4] Another advanced development is to deliver siRNA byPEGylated cyclodextrin molecules [5] They were released bydissociation of the complexes in lysosome Particularly themost commonly used drug delivery system is the polymericmaterials [6]The biblock copolymers tend to formmicelle inthe presence of drug molecules Therefore drug can be easilyloaded into the core of micelle [7] However being usefuldrug nanocarriers it is necessary to consider their toxicitybiocompatibility and stability in a cellular environment Itis well-known that most of the nanoparticles are toxic they

Hindawi Publishing CorporationJournal of NanomaterialsVolume 2015 Article ID 765492 21 pageshttpdxdoiorg1011552015765492

2 Journal of Nanomaterials

may induce cytotoxicity in living systems Heat triggered-release of drug molecules in a cellular environment is notappreciated because other healthy cells may also be affectedIn addition the efficiency and selectivity of drug loadingin polymeric micelles is also highly limited Therefore todesign new materials as drug carriers these carriers shouldhave a capability of drug incorporation and controlled releasein a highly effective way They should also be highly stableand biocompatible in a specific cellular environment It isalso necessary for them to target particular areas and carrymultifunction in order to enhance the delivery efficiency

Indeed developing novel biocompatible and multifunc-tional nanocarriers remains a key challenge for targeted drugdelivery The rapidly emerging DNA nanotechnology beganwith pioneer Seemanrsquos hypothesis that DNA not only cancarry genetic information but also can be used as molecularorganizer to create well-designed and controllable nanoma-terials for applications in materials science nanotechnologyand biology [8 9] AsDNAhas a simple and robustmolecularrecognition rule of adenine to thymine (A-T) and guanine tocytosine (G-C) pairings two complementary single-strandedDNA hybridize to form a double helix with predictableand programmable interactions The structural features ofnucleic acids form the basis of constructing a wide varietyof well-ordered DNA nanoarchitectures with well-definedshapes and sizes in addition to controllable permeabilityand flexibility [10 11] This DNA nanotechnology offersnew opportunities for the construction of complex DNAstructures in different dimensions More importantly self-assembled DNA nanostructures can be easily functionalizedto construct artificial functional systems for multipurposesApparently scientists envision artificial DNA-based nanos-tructures as tools for drug loading and in vivo targeteddelivery because of their potential of selective encapsulationand stimuli-triggered release of cargo

In this review article we concentrate on a new-comerof drug delivery carriers based on self-assembled DNAnanostructures We will demonstrate the power and promiseof DNA as a scaffold to create DNA nanostructures withprecise geometry and versatile functionality Their structuralstability in physiological conditions and internalization willbe briefly described Different cargo loadingmechanisms andtheir control release via external stimuli will be summarizedin detail As a new-comer in drug delivery system studiesof intracellular behaviorsfunctions of drug loaded DNAnanocarriers and their interactions in specific intracellularcompartments in vitro or in vivo will also be discussedSome concluding remarks will try to ascertain what the nextchallenges and outlook of this exciting research area could be

2 DNA NanotechnologyTo begin with we first briefly introduce the history and themost updated status of DNAnanotechnologyThe innovationof the field of DNA nanotechnology was first demonstratedby Seeman in the early 1980s [12] Taking advantage ofself-recognition property of DNA his group designed andconstructed modified Holliday junctions to convert one-dimensional DNA strands into branched DNA tiles with

sticky ends at the edges (Figure 1(a)) These short single-stranded units provide toeholds for further assembly of 2D-structures [13] Since then the structural role of DNA iswidely well-recognized and extensively explored Howeverthese assembly approaches did not offer rigid junctions withwell-defined angles and geometry of the final structures Toovercome these drawbacks researchers started to developadvanced rigid junctions including multicrossover [14ndash16]cross-shaped tile with arms [17] DNA tensegrity triangle[18] and parallelogram DNA tile (Figure 1(b)) [19] Withsuch unprecedented talent to construct DNA-based architec-tures highly ordered 2D-DNA surfaces with programmablearrangement and a large variety of three-dimensional poly-hedral structures were successfully assembled via sticky-end cohesion among those building blocks [20ndash22] Never-theless these tile-based assemblies have certain limitationsFor example it is difficult to control the size of resultingstructures An exact stoichiometric and a high purity controlof individual DNA fragments are still problematic for theassembly of large and complex nanostructures

Another creation in DNA nanotechnology was madeby Rothemund in 2006 [23] He invented scaffolded DNAorigami which successfully offered high complexity andversatility in DNA assembly In DNA origami a long pieceof single-stranded DNA from theM13 circular bacteriophagegenome is folded with itself into a desired pattern withthe assistance of short staple strands (Figure 1(c)) [24 25]Typical examples consist of nonperiodic 2D-structures suchas a map of the Americas stars smiley faces and otherdeliberately well-designed patterns [26 27] In this approachthe relative stoichiometric ratio on different staple strandsto a single DNA scaffold is not highly restricted Moreimportantly DNA origami is a versatile and simple one-potassembly to generate nanostructures with complex shapesof predefined dimensions as compared to the conventionalcrossover approach [28 29] In an advanced developmentKostiainenrsquos group has recently demonstrated the opticalcontrol of the DNA origami formation and release [30]Although DNA was used as the only component to guide theDNA assembly in tile-based assembly or DNA origami thisresulted in fully double-stranded and DNA-dense structures

An alternative approach to building DNA nanostructureis to bring together the programmability of DNA with func-tional and structural diversities offered by supramolecularchemistry [31]This new emerging area inDNAnanotechnol-ogy involves the insertion of synthetic molecules into DNAstrand to alter its hybridization and control the assemblyoutcome (Figure 1(d)) By conjugating synthetic moleculesat the insertion points of a DNA strand typical linearDNA duplexes can be oriented and hybridized relative toone another in a controlled manner This supramolecularDNA assembly combines the diverse structural features ofmolecules and their functionalities such as luminescenceredox magnetic and catalytic properties to generate discretewell-defined structures

Taking advantages of synthetic molecules as rigid junc-tions this can reduce the amount of DNA strands neededfor the structural definition as compared to the previoustwo methods For example Sleimanrsquos group have successfully

Journal of Nanomaterials 3

Holliday junction

H

998400H

998400H

998400HV

998400V

998400V998400V

H

H

V V

(a)TCTGATGT

ACTACA

GAGCAGCCCGTCGG

TGTACGGACATGCC

CCGTACA

GGCATGT CCGTACA

GGCATGT

TCTGATGT

ACTACA

GGCTGC

CCGACGAG

GGCTGC

CCGACGAG

ACATCATGTAGTCT

(b)

(c)

Syntheticmolecule

(d)

Figure 1 Examples of self-assembled DNA nanostructures (a) A lattice is formed by hybridization of the sticky ends of a Holliday junction(b) multistranded junction structures and crossover structures including double-crossover structure cross-shaped tile with four arms DNAtensegrity triangle and parallelogram DNA tile (c) the principle of DNA origami and the design of 2D origami formed smiling face and star(d) sequential self-assembly of hexagonal-shaped DNA nanostructure via supramolecular DNA assembly

developed DNA-conjugated m-terphenyl-based organic ver-tices for modular construction of cyclic polygons a library ofDNA polyhedral structures and nanotubes with good controlover their geometry [32] dimension [33] and flexibility [34]Besides the organic insertions other important self-assemblystrategies take advantages of transition metal- ligand- lipid-and block copolymer-based environments [35ndash37]

3 Stability of Self-AssembledDNA Nanostructures

Among various DNA assemblies three-dimensional DNAnanostructures hold promise to be the universal nanocarriers

for smart and targeted drug delivery In contrast to 1D or2D DNA structures the power of self-assembled 3D DNAnanostructures lies in their excellent stability and biocom-patibility high drug loading capability and passive deliveryinto live cells They also possess fine control over geometryprecise and monodisperse dimensions positioning of guestmolecules stimuli-responsive switching of structure andtriggered-release of cargos Typical examples of drug deliverysystems based on 3D DNA nanostructures [38 39] includetetrahedron icosahedron hexagonal barrel nanotube DNAorigami box [40] nanorobot and nanocage

To be employable as a drug carrier system in mammalsDNA nanostructures must meet several important criteria

4 Journal of Nanomaterials

Table 1 Stability of different DNA nanostructures

Linear dsDNA CpGbearingDNA tetrahedral nanostructure

A 3D multilayer rectangularparallelepiped structure

CG

CG

CG

CG

Description of the structure Normal linear DNA strand withDdeI restriction site

It is made up of four 55-merstrands extended with the CpG

sequence and a 7-meroligothymine spacer

A 3D multilayer rectangularparallelepiped structure (8 helixtimes 8 helix square lattice with

dimensions of 16 nm times 16 nm times30 nm)

Incubation temperature 37∘C 25∘C

Medium 10 FBS 50 non-heat-inactivated fetalbovine serum (FBS) Cell lysate

Decay time Decay after 08 h Start decaying after 4 h but stillnot completely decayed after 24 h Still remains stable after 12 h

Citation [41] [43] [44]

(1) they have to be stable and intact in both extracellular andintracellular environments particularly stable long enoughin the cytoplasm of cells to perform their predefined tasks(2) they should not have toxic effect in mammals and (3)the cellular immune system in mammals should toleratethe nanometer-scale DNA nanocarrier systems Thus farseveral research groups have put efforts on the stabilitystudies of DNA constructs Bermudezrsquos group indicatedthat oligonucleotide-based tetrahedral made from branchjunctions exhibit a strong resistance to enzymatic digestioncompared to the linear counterparts in terms of their decaytime constants (Table 1) [41] The reason behind this wouldhighly be due to the steric hindrance effect Since the endonu-cleases initially bind to the DNA nonspecifically with a lowaffinity and then follow by diffusion along the strands Thesteric hindrance introduced by three-dimensional tetrahe-dron would reduce the effective binding of enzymes to DNAand then inhibit DNA cleavage no matter if the enzyme actsspecifically or nonspecifically Furthermore shorter sequenceor smaller size of DNA complex can enhance the resistancetowards various nucleases as they are more difficult to bendand possibly have higher steric hindrance for the action ofthe enzymes Walsh and coworkers have demonstrated thefirst example of 3D DNA nanostructure which can enter livemammalian cells effectively with or without the help of atransfection reagent [42] They stay intact for up to 48 h incytoplasm In a recent study by Li et al they have modifiedthe tetrahedral with CpG oligonucleotides which have beenconfirmed to be taken up by macrophage RAW2647 cellseffectively (Table 1) [43]

Regarding scaffold DNA origami Mei and coworkersdemonstrated that different shapes of DNA origami nanos-tructures are stable and remain intact for 12 h after exposing

to cell lysates of various cell lines and can be easily puri-fied from lysate mixtures in contrast to single-strandedor duplex DNA (Table 1) [44] They are not accessible tovarious DNAzymes due to negatively charged large andrigid origami structures Their superior structural integrityand versatile functionality are highly preserved in relationto conventional oligonucleotides validating their use forvarious biological applications Subsequently a further studycarried out by Dietzrsquo group tested the enzymatic digestion ofDNAorigami structures [45]They are fully exposed to a largevariety of endonucleases including DNase T7 exonucleaseT7 endonuclease Msel restriction endonuclease Lambdaexonuclease and Escherichia coli exonuclease These resultsindicated that they are highly stable at 37∘C towards degra-dation as compared to duplex plasmid oligonucleotidesMore recently Schuller and his coworkers reported that CpGoligonucleotides-decorated DNA origami tubes amplify astrong immune response which are completely dependent onTLR9 stimulation in mammalian spleen cell [46]

To further optimize DNA structures in regard to enzy-matic digestion resistance Sleimanrsquos group has modified 3DDNA nanostructures using a number of chemical strategiesThey found that simple chemical modification to both endsof DNA oligos with hexanediol and hexaethylene glycol inself-assembledDNAprismatic cage or site-specific hybridiza-tion of DNA-block copolymer chains to 3D DNA scaffoldwould dramatically enhance its nuclease resistance underfetal bovine serum condition (Table 2) [47] These studiescould provide guidelines for decoration of DNA nanostruc-tures with simple chemistry modification and allow impart-ing momentous stabilization towards nuclease degradationMeanwhile the same group also demonstrated that creationof DNA nanotubes with a template generated by rolling circle

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 3: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

2 Journal of Nanomaterials

may induce cytotoxicity in living systems Heat triggered-release of drug molecules in a cellular environment is notappreciated because other healthy cells may also be affectedIn addition the efficiency and selectivity of drug loadingin polymeric micelles is also highly limited Therefore todesign new materials as drug carriers these carriers shouldhave a capability of drug incorporation and controlled releasein a highly effective way They should also be highly stableand biocompatible in a specific cellular environment It isalso necessary for them to target particular areas and carrymultifunction in order to enhance the delivery efficiency

Indeed developing novel biocompatible and multifunc-tional nanocarriers remains a key challenge for targeted drugdelivery The rapidly emerging DNA nanotechnology beganwith pioneer Seemanrsquos hypothesis that DNA not only cancarry genetic information but also can be used as molecularorganizer to create well-designed and controllable nanoma-terials for applications in materials science nanotechnologyand biology [8 9] AsDNAhas a simple and robustmolecularrecognition rule of adenine to thymine (A-T) and guanine tocytosine (G-C) pairings two complementary single-strandedDNA hybridize to form a double helix with predictableand programmable interactions The structural features ofnucleic acids form the basis of constructing a wide varietyof well-ordered DNA nanoarchitectures with well-definedshapes and sizes in addition to controllable permeabilityand flexibility [10 11] This DNA nanotechnology offersnew opportunities for the construction of complex DNAstructures in different dimensions More importantly self-assembled DNA nanostructures can be easily functionalizedto construct artificial functional systems for multipurposesApparently scientists envision artificial DNA-based nanos-tructures as tools for drug loading and in vivo targeteddelivery because of their potential of selective encapsulationand stimuli-triggered release of cargo

In this review article we concentrate on a new-comerof drug delivery carriers based on self-assembled DNAnanostructures We will demonstrate the power and promiseof DNA as a scaffold to create DNA nanostructures withprecise geometry and versatile functionality Their structuralstability in physiological conditions and internalization willbe briefly described Different cargo loadingmechanisms andtheir control release via external stimuli will be summarizedin detail As a new-comer in drug delivery system studiesof intracellular behaviorsfunctions of drug loaded DNAnanocarriers and their interactions in specific intracellularcompartments in vitro or in vivo will also be discussedSome concluding remarks will try to ascertain what the nextchallenges and outlook of this exciting research area could be

2 DNA NanotechnologyTo begin with we first briefly introduce the history and themost updated status of DNAnanotechnologyThe innovationof the field of DNA nanotechnology was first demonstratedby Seeman in the early 1980s [12] Taking advantage ofself-recognition property of DNA his group designed andconstructed modified Holliday junctions to convert one-dimensional DNA strands into branched DNA tiles with

sticky ends at the edges (Figure 1(a)) These short single-stranded units provide toeholds for further assembly of 2D-structures [13] Since then the structural role of DNA iswidely well-recognized and extensively explored Howeverthese assembly approaches did not offer rigid junctions withwell-defined angles and geometry of the final structures Toovercome these drawbacks researchers started to developadvanced rigid junctions including multicrossover [14ndash16]cross-shaped tile with arms [17] DNA tensegrity triangle[18] and parallelogram DNA tile (Figure 1(b)) [19] Withsuch unprecedented talent to construct DNA-based architec-tures highly ordered 2D-DNA surfaces with programmablearrangement and a large variety of three-dimensional poly-hedral structures were successfully assembled via sticky-end cohesion among those building blocks [20ndash22] Never-theless these tile-based assemblies have certain limitationsFor example it is difficult to control the size of resultingstructures An exact stoichiometric and a high purity controlof individual DNA fragments are still problematic for theassembly of large and complex nanostructures

Another creation in DNA nanotechnology was madeby Rothemund in 2006 [23] He invented scaffolded DNAorigami which successfully offered high complexity andversatility in DNA assembly In DNA origami a long pieceof single-stranded DNA from theM13 circular bacteriophagegenome is folded with itself into a desired pattern withthe assistance of short staple strands (Figure 1(c)) [24 25]Typical examples consist of nonperiodic 2D-structures suchas a map of the Americas stars smiley faces and otherdeliberately well-designed patterns [26 27] In this approachthe relative stoichiometric ratio on different staple strandsto a single DNA scaffold is not highly restricted Moreimportantly DNA origami is a versatile and simple one-potassembly to generate nanostructures with complex shapesof predefined dimensions as compared to the conventionalcrossover approach [28 29] In an advanced developmentKostiainenrsquos group has recently demonstrated the opticalcontrol of the DNA origami formation and release [30]Although DNA was used as the only component to guide theDNA assembly in tile-based assembly or DNA origami thisresulted in fully double-stranded and DNA-dense structures

An alternative approach to building DNA nanostructureis to bring together the programmability of DNA with func-tional and structural diversities offered by supramolecularchemistry [31]This new emerging area inDNAnanotechnol-ogy involves the insertion of synthetic molecules into DNAstrand to alter its hybridization and control the assemblyoutcome (Figure 1(d)) By conjugating synthetic moleculesat the insertion points of a DNA strand typical linearDNA duplexes can be oriented and hybridized relative toone another in a controlled manner This supramolecularDNA assembly combines the diverse structural features ofmolecules and their functionalities such as luminescenceredox magnetic and catalytic properties to generate discretewell-defined structures

Taking advantages of synthetic molecules as rigid junc-tions this can reduce the amount of DNA strands neededfor the structural definition as compared to the previoustwo methods For example Sleimanrsquos group have successfully

Journal of Nanomaterials 3

Holliday junction

H

998400H

998400H

998400HV

998400V

998400V998400V

H

H

V V

(a)TCTGATGT

ACTACA

GAGCAGCCCGTCGG

TGTACGGACATGCC

CCGTACA

GGCATGT CCGTACA

GGCATGT

TCTGATGT

ACTACA

GGCTGC

CCGACGAG

GGCTGC

CCGACGAG

ACATCATGTAGTCT

(b)

(c)

Syntheticmolecule

(d)

Figure 1 Examples of self-assembled DNA nanostructures (a) A lattice is formed by hybridization of the sticky ends of a Holliday junction(b) multistranded junction structures and crossover structures including double-crossover structure cross-shaped tile with four arms DNAtensegrity triangle and parallelogram DNA tile (c) the principle of DNA origami and the design of 2D origami formed smiling face and star(d) sequential self-assembly of hexagonal-shaped DNA nanostructure via supramolecular DNA assembly

developed DNA-conjugated m-terphenyl-based organic ver-tices for modular construction of cyclic polygons a library ofDNA polyhedral structures and nanotubes with good controlover their geometry [32] dimension [33] and flexibility [34]Besides the organic insertions other important self-assemblystrategies take advantages of transition metal- ligand- lipid-and block copolymer-based environments [35ndash37]

3 Stability of Self-AssembledDNA Nanostructures

Among various DNA assemblies three-dimensional DNAnanostructures hold promise to be the universal nanocarriers

for smart and targeted drug delivery In contrast to 1D or2D DNA structures the power of self-assembled 3D DNAnanostructures lies in their excellent stability and biocom-patibility high drug loading capability and passive deliveryinto live cells They also possess fine control over geometryprecise and monodisperse dimensions positioning of guestmolecules stimuli-responsive switching of structure andtriggered-release of cargos Typical examples of drug deliverysystems based on 3D DNA nanostructures [38 39] includetetrahedron icosahedron hexagonal barrel nanotube DNAorigami box [40] nanorobot and nanocage

To be employable as a drug carrier system in mammalsDNA nanostructures must meet several important criteria

4 Journal of Nanomaterials

Table 1 Stability of different DNA nanostructures

Linear dsDNA CpGbearingDNA tetrahedral nanostructure

A 3D multilayer rectangularparallelepiped structure

CG

CG

CG

CG

Description of the structure Normal linear DNA strand withDdeI restriction site

It is made up of four 55-merstrands extended with the CpG

sequence and a 7-meroligothymine spacer

A 3D multilayer rectangularparallelepiped structure (8 helixtimes 8 helix square lattice with

dimensions of 16 nm times 16 nm times30 nm)

Incubation temperature 37∘C 25∘C

Medium 10 FBS 50 non-heat-inactivated fetalbovine serum (FBS) Cell lysate

Decay time Decay after 08 h Start decaying after 4 h but stillnot completely decayed after 24 h Still remains stable after 12 h

Citation [41] [43] [44]

(1) they have to be stable and intact in both extracellular andintracellular environments particularly stable long enoughin the cytoplasm of cells to perform their predefined tasks(2) they should not have toxic effect in mammals and (3)the cellular immune system in mammals should toleratethe nanometer-scale DNA nanocarrier systems Thus farseveral research groups have put efforts on the stabilitystudies of DNA constructs Bermudezrsquos group indicatedthat oligonucleotide-based tetrahedral made from branchjunctions exhibit a strong resistance to enzymatic digestioncompared to the linear counterparts in terms of their decaytime constants (Table 1) [41] The reason behind this wouldhighly be due to the steric hindrance effect Since the endonu-cleases initially bind to the DNA nonspecifically with a lowaffinity and then follow by diffusion along the strands Thesteric hindrance introduced by three-dimensional tetrahe-dron would reduce the effective binding of enzymes to DNAand then inhibit DNA cleavage no matter if the enzyme actsspecifically or nonspecifically Furthermore shorter sequenceor smaller size of DNA complex can enhance the resistancetowards various nucleases as they are more difficult to bendand possibly have higher steric hindrance for the action ofthe enzymes Walsh and coworkers have demonstrated thefirst example of 3D DNA nanostructure which can enter livemammalian cells effectively with or without the help of atransfection reagent [42] They stay intact for up to 48 h incytoplasm In a recent study by Li et al they have modifiedthe tetrahedral with CpG oligonucleotides which have beenconfirmed to be taken up by macrophage RAW2647 cellseffectively (Table 1) [43]

Regarding scaffold DNA origami Mei and coworkersdemonstrated that different shapes of DNA origami nanos-tructures are stable and remain intact for 12 h after exposing

to cell lysates of various cell lines and can be easily puri-fied from lysate mixtures in contrast to single-strandedor duplex DNA (Table 1) [44] They are not accessible tovarious DNAzymes due to negatively charged large andrigid origami structures Their superior structural integrityand versatile functionality are highly preserved in relationto conventional oligonucleotides validating their use forvarious biological applications Subsequently a further studycarried out by Dietzrsquo group tested the enzymatic digestion ofDNAorigami structures [45]They are fully exposed to a largevariety of endonucleases including DNase T7 exonucleaseT7 endonuclease Msel restriction endonuclease Lambdaexonuclease and Escherichia coli exonuclease These resultsindicated that they are highly stable at 37∘C towards degra-dation as compared to duplex plasmid oligonucleotidesMore recently Schuller and his coworkers reported that CpGoligonucleotides-decorated DNA origami tubes amplify astrong immune response which are completely dependent onTLR9 stimulation in mammalian spleen cell [46]

To further optimize DNA structures in regard to enzy-matic digestion resistance Sleimanrsquos group has modified 3DDNA nanostructures using a number of chemical strategiesThey found that simple chemical modification to both endsof DNA oligos with hexanediol and hexaethylene glycol inself-assembledDNAprismatic cage or site-specific hybridiza-tion of DNA-block copolymer chains to 3D DNA scaffoldwould dramatically enhance its nuclease resistance underfetal bovine serum condition (Table 2) [47] These studiescould provide guidelines for decoration of DNA nanostruc-tures with simple chemistry modification and allow impart-ing momentous stabilization towards nuclease degradationMeanwhile the same group also demonstrated that creationof DNA nanotubes with a template generated by rolling circle

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 4: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 3

Holliday junction

H

998400H

998400H

998400HV

998400V

998400V998400V

H

H

V V

(a)TCTGATGT

ACTACA

GAGCAGCCCGTCGG

TGTACGGACATGCC

CCGTACA

GGCATGT CCGTACA

GGCATGT

TCTGATGT

ACTACA

GGCTGC

CCGACGAG

GGCTGC

CCGACGAG

ACATCATGTAGTCT

(b)

(c)

Syntheticmolecule

(d)

Figure 1 Examples of self-assembled DNA nanostructures (a) A lattice is formed by hybridization of the sticky ends of a Holliday junction(b) multistranded junction structures and crossover structures including double-crossover structure cross-shaped tile with four arms DNAtensegrity triangle and parallelogram DNA tile (c) the principle of DNA origami and the design of 2D origami formed smiling face and star(d) sequential self-assembly of hexagonal-shaped DNA nanostructure via supramolecular DNA assembly

developed DNA-conjugated m-terphenyl-based organic ver-tices for modular construction of cyclic polygons a library ofDNA polyhedral structures and nanotubes with good controlover their geometry [32] dimension [33] and flexibility [34]Besides the organic insertions other important self-assemblystrategies take advantages of transition metal- ligand- lipid-and block copolymer-based environments [35ndash37]

3 Stability of Self-AssembledDNA Nanostructures

Among various DNA assemblies three-dimensional DNAnanostructures hold promise to be the universal nanocarriers

for smart and targeted drug delivery In contrast to 1D or2D DNA structures the power of self-assembled 3D DNAnanostructures lies in their excellent stability and biocom-patibility high drug loading capability and passive deliveryinto live cells They also possess fine control over geometryprecise and monodisperse dimensions positioning of guestmolecules stimuli-responsive switching of structure andtriggered-release of cargos Typical examples of drug deliverysystems based on 3D DNA nanostructures [38 39] includetetrahedron icosahedron hexagonal barrel nanotube DNAorigami box [40] nanorobot and nanocage

To be employable as a drug carrier system in mammalsDNA nanostructures must meet several important criteria

4 Journal of Nanomaterials

Table 1 Stability of different DNA nanostructures

Linear dsDNA CpGbearingDNA tetrahedral nanostructure

A 3D multilayer rectangularparallelepiped structure

CG

CG

CG

CG

Description of the structure Normal linear DNA strand withDdeI restriction site

It is made up of four 55-merstrands extended with the CpG

sequence and a 7-meroligothymine spacer

A 3D multilayer rectangularparallelepiped structure (8 helixtimes 8 helix square lattice with

dimensions of 16 nm times 16 nm times30 nm)

Incubation temperature 37∘C 25∘C

Medium 10 FBS 50 non-heat-inactivated fetalbovine serum (FBS) Cell lysate

Decay time Decay after 08 h Start decaying after 4 h but stillnot completely decayed after 24 h Still remains stable after 12 h

Citation [41] [43] [44]

(1) they have to be stable and intact in both extracellular andintracellular environments particularly stable long enoughin the cytoplasm of cells to perform their predefined tasks(2) they should not have toxic effect in mammals and (3)the cellular immune system in mammals should toleratethe nanometer-scale DNA nanocarrier systems Thus farseveral research groups have put efforts on the stabilitystudies of DNA constructs Bermudezrsquos group indicatedthat oligonucleotide-based tetrahedral made from branchjunctions exhibit a strong resistance to enzymatic digestioncompared to the linear counterparts in terms of their decaytime constants (Table 1) [41] The reason behind this wouldhighly be due to the steric hindrance effect Since the endonu-cleases initially bind to the DNA nonspecifically with a lowaffinity and then follow by diffusion along the strands Thesteric hindrance introduced by three-dimensional tetrahe-dron would reduce the effective binding of enzymes to DNAand then inhibit DNA cleavage no matter if the enzyme actsspecifically or nonspecifically Furthermore shorter sequenceor smaller size of DNA complex can enhance the resistancetowards various nucleases as they are more difficult to bendand possibly have higher steric hindrance for the action ofthe enzymes Walsh and coworkers have demonstrated thefirst example of 3D DNA nanostructure which can enter livemammalian cells effectively with or without the help of atransfection reagent [42] They stay intact for up to 48 h incytoplasm In a recent study by Li et al they have modifiedthe tetrahedral with CpG oligonucleotides which have beenconfirmed to be taken up by macrophage RAW2647 cellseffectively (Table 1) [43]

Regarding scaffold DNA origami Mei and coworkersdemonstrated that different shapes of DNA origami nanos-tructures are stable and remain intact for 12 h after exposing

to cell lysates of various cell lines and can be easily puri-fied from lysate mixtures in contrast to single-strandedor duplex DNA (Table 1) [44] They are not accessible tovarious DNAzymes due to negatively charged large andrigid origami structures Their superior structural integrityand versatile functionality are highly preserved in relationto conventional oligonucleotides validating their use forvarious biological applications Subsequently a further studycarried out by Dietzrsquo group tested the enzymatic digestion ofDNAorigami structures [45]They are fully exposed to a largevariety of endonucleases including DNase T7 exonucleaseT7 endonuclease Msel restriction endonuclease Lambdaexonuclease and Escherichia coli exonuclease These resultsindicated that they are highly stable at 37∘C towards degra-dation as compared to duplex plasmid oligonucleotidesMore recently Schuller and his coworkers reported that CpGoligonucleotides-decorated DNA origami tubes amplify astrong immune response which are completely dependent onTLR9 stimulation in mammalian spleen cell [46]

To further optimize DNA structures in regard to enzy-matic digestion resistance Sleimanrsquos group has modified 3DDNA nanostructures using a number of chemical strategiesThey found that simple chemical modification to both endsof DNA oligos with hexanediol and hexaethylene glycol inself-assembledDNAprismatic cage or site-specific hybridiza-tion of DNA-block copolymer chains to 3D DNA scaffoldwould dramatically enhance its nuclease resistance underfetal bovine serum condition (Table 2) [47] These studiescould provide guidelines for decoration of DNA nanostruc-tures with simple chemistry modification and allow impart-ing momentous stabilization towards nuclease degradationMeanwhile the same group also demonstrated that creationof DNA nanotubes with a template generated by rolling circle

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 5: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

4 Journal of Nanomaterials

Table 1 Stability of different DNA nanostructures

Linear dsDNA CpGbearingDNA tetrahedral nanostructure

A 3D multilayer rectangularparallelepiped structure

CG

CG

CG

CG

Description of the structure Normal linear DNA strand withDdeI restriction site

It is made up of four 55-merstrands extended with the CpG

sequence and a 7-meroligothymine spacer

A 3D multilayer rectangularparallelepiped structure (8 helixtimes 8 helix square lattice with

dimensions of 16 nm times 16 nm times30 nm)

Incubation temperature 37∘C 25∘C

Medium 10 FBS 50 non-heat-inactivated fetalbovine serum (FBS) Cell lysate

Decay time Decay after 08 h Start decaying after 4 h but stillnot completely decayed after 24 h Still remains stable after 12 h

Citation [41] [43] [44]

(1) they have to be stable and intact in both extracellular andintracellular environments particularly stable long enoughin the cytoplasm of cells to perform their predefined tasks(2) they should not have toxic effect in mammals and (3)the cellular immune system in mammals should toleratethe nanometer-scale DNA nanocarrier systems Thus farseveral research groups have put efforts on the stabilitystudies of DNA constructs Bermudezrsquos group indicatedthat oligonucleotide-based tetrahedral made from branchjunctions exhibit a strong resistance to enzymatic digestioncompared to the linear counterparts in terms of their decaytime constants (Table 1) [41] The reason behind this wouldhighly be due to the steric hindrance effect Since the endonu-cleases initially bind to the DNA nonspecifically with a lowaffinity and then follow by diffusion along the strands Thesteric hindrance introduced by three-dimensional tetrahe-dron would reduce the effective binding of enzymes to DNAand then inhibit DNA cleavage no matter if the enzyme actsspecifically or nonspecifically Furthermore shorter sequenceor smaller size of DNA complex can enhance the resistancetowards various nucleases as they are more difficult to bendand possibly have higher steric hindrance for the action ofthe enzymes Walsh and coworkers have demonstrated thefirst example of 3D DNA nanostructure which can enter livemammalian cells effectively with or without the help of atransfection reagent [42] They stay intact for up to 48 h incytoplasm In a recent study by Li et al they have modifiedthe tetrahedral with CpG oligonucleotides which have beenconfirmed to be taken up by macrophage RAW2647 cellseffectively (Table 1) [43]

Regarding scaffold DNA origami Mei and coworkersdemonstrated that different shapes of DNA origami nanos-tructures are stable and remain intact for 12 h after exposing

to cell lysates of various cell lines and can be easily puri-fied from lysate mixtures in contrast to single-strandedor duplex DNA (Table 1) [44] They are not accessible tovarious DNAzymes due to negatively charged large andrigid origami structures Their superior structural integrityand versatile functionality are highly preserved in relationto conventional oligonucleotides validating their use forvarious biological applications Subsequently a further studycarried out by Dietzrsquo group tested the enzymatic digestion ofDNAorigami structures [45]They are fully exposed to a largevariety of endonucleases including DNase T7 exonucleaseT7 endonuclease Msel restriction endonuclease Lambdaexonuclease and Escherichia coli exonuclease These resultsindicated that they are highly stable at 37∘C towards degra-dation as compared to duplex plasmid oligonucleotidesMore recently Schuller and his coworkers reported that CpGoligonucleotides-decorated DNA origami tubes amplify astrong immune response which are completely dependent onTLR9 stimulation in mammalian spleen cell [46]

To further optimize DNA structures in regard to enzy-matic digestion resistance Sleimanrsquos group has modified 3DDNA nanostructures using a number of chemical strategiesThey found that simple chemical modification to both endsof DNA oligos with hexanediol and hexaethylene glycol inself-assembledDNAprismatic cage or site-specific hybridiza-tion of DNA-block copolymer chains to 3D DNA scaffoldwould dramatically enhance its nuclease resistance underfetal bovine serum condition (Table 2) [47] These studiescould provide guidelines for decoration of DNA nanostruc-tures with simple chemistry modification and allow impart-ing momentous stabilization towards nuclease degradationMeanwhile the same group also demonstrated that creationof DNA nanotubes with a template generated by rolling circle

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 6: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 5

Table 2 Stability of modified DNA nanostructures generated from supramolecular DNA assembly

Triangular prism1 Triangular prism2 Nanotube RCA-nanotube

Triangular prism(TP)

Description ofthe structure

Made up of three96-mer strandswith 20 bp edges

Made up of three96-mer hexaethylene

glycol (HEG)modified strands with

20 bp edges

Triangular prism built up bysmall unit with short linking

DNA strand

Connect small triangular prismunits with RCA synthesized

DNA strand

Incubation temperature 37∘C 37∘C medium 10 FBS 10 FBS 10 FBS 10 FBSDecay time 18 h 62 h 11 h 35 hCitation [47] [47] [32 48] [48]

amplification (RCA) results in increased stability towardsnuclease degradation as compared to their previous nanotubedesign (Table 2) [48] On the other hand the high density ofDNA and aspect ratio of the RCA-templated DNA nanotubesoffer a greater cell penetration ability over normal DNAoligos Such enhanced cellular stability and nuclease suscep-tibility are the key requirements for DNA nanostructures toact as delivery carriers or vehicles

To modulate the stability and uptake profile of self-assembled DNA nanocube Sleimanrsquos group recently deco-rated their DNA cubes with hydrophobic (dodecane alkylC12) or hydrophilic (hexaethylene glycol HEG) dendriticDNA chains [49] or block copolymers on the edges [50]They found that all of the integrating dendritic DNA chainswere facing outward as confirmed by a larger hydrody-namic radius from dynamic light scattering (DLS) study andlower mobility band on gel electrophoresis In addition thischemical modification would allow enhancing their cellularstability with a longer half-life as compared to the blunt-ended nanocubes More importantly they found that thehydrophobic chains on the cube favor rapid and increasedcellular uptake while the hydrophilic chains favor slow andcontinuous internalization

4 Cargo Loading and Cellular Delivery

In response to the well-defined and highly programmableproperties of DNA-based nanostructures precise control

of positioning of cargo molecules in DNA nano-objects ishighly possible This valuable property is hardly attainablewith inorganic or organic nanomaterials In general cargomolecules can be loaded via different strategies such ascovalent linkage nucleic acid base-pairing biotin-avidininteraction intercalation aptamer-target interaction DNA-protein interaction and encapsulation

41 Covalent Linkage To deliver the cargo with the aid ofDNA nanostructures some of the cargos can form covalentbonds with DNA strand in the presence of some molecularlinkers Sleimanrsquos and Maorsquos groups have shown that self-assembled DNA nanotubes act as carriers to deliver cyaninefluorescent dyes into human cancer cells [48 51] In Maostudy Cy3 is covalently conjugated to some of the nucleicacid strands at their 51015840 ends via a well-established N-hydroxysuccinimide (NHS) chemistry Cy3-functionalizedDNA nanotubes were formed by mixing DNA strands withand without Cy3 molecules after a heart-cool cycle Flu-orescent dyes are the most commonly used model cargofor targeted delivery because they can easily be visualizedand traced under various fluorescence microscopes Takingadvantage of automatic solid-phase DNA synthesis a widerange of fluorescent probes can be readily coupled andlabeled on DNA stands Withwithout the help of targetingmoieties these structures could be internalized by tumorcells The fluorescence of the dyes could be localized withfluorescent microscopy confirming the presence of DNA

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 7: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

6 Journal of Nanomaterials

(a)

DNA-AuNP

Tail-TET

Tail-OCT

Tail-ICO

AuNPTET

AuNPOCT

AuNPICO

(b)

Figure 2 (a) Different kinds of antibodies have been tagged on the nanorobot and it can identify different antigens on different cells (b)Thecomplementary strand is incorporated inside the cavity of the nanocage for encapsulation of gold

nanoassemblies in cells Moreover we are able to preciselycontrol the numbers and positions of these fluorescent cargossuch that multiple fluorophores can be labeled on a singleDNA nanostructure [42 52]

42 Nucleic Acid Base-Pairing Hybridization of cargo-consisting of single-stranded nucleic acids offers an alter-native strategy for site-specific loading of cargos Thenanorobots produced by Churchrsquos group have been chem-ically modified via covalent attachment of 15-base ssDNAlinkers as loading sites to the 51015840 ends of payloads (Figure 2(a))[53] In this structure twelve loading sites were gener-ated Subsequently two types of cargo linkers have beenprepared in the following ways gold nanoparticles cova-lently conjugated to 51015840-thiol-functionalized DNA linkersand Fabrsquo antibodies were covalently conjugated to 51015840-amine-functionalized DNA linkers Mixing the cargo linkers andthe nanorobot in aqueous buffer the staple strands with 31015840extensions localized at the loading sites hybridized with thecomplementary sequences of cargo linkers Eventually twodifferent types of payload molecules are loaded successfullyper robot In their design different Fabrsquo antibody fragmentswere bounded covalently to the amine-modified linkersThey found that the antibodies were recognized by certaincell-surface receptors and thus inhibited the growth of thetargeted cells In addition generality of using these barrelstructures as carrier is highly possible because a decrease in Tcell activation activity that was observed when Fab fragmentstargeted to human CD3 and flagellin were loaded on thesehexagonal barrel structures

Maorsquos group has designed a series of symmetric DNApolyhedral structures consisting of two unpaired ss DNAtails sticking out on each edge (Figure 2(b)) [54] Whenmixing the gold nanoparticles functionalized with DNA

strands (DNA-AuNPs) the DNA-AuNPs are swallowed intothe polyhedral structures governed by nucleic acid basepairing between the ssDNA tail on the DNA polyhedralstructures and the complementaryDNA strands immobilizedon AuNPs The size and number of guest molecules trappedby these DNA polyhedra highly depend on their internalvolumes

An alternative molecular cargo drawing attention isRNA interference (RNAi) It becomes a powerful therapeuticagent to knock down the gene expression inducing genesilencing Small interfering RNAs (siRNAs) are chemicallysynthesized nucleic acids with specific sequences which bindto their complementary mRNA molecules and thus inhibitthe corresponding protein synthesis leading to targeted geneknockdown By choosing the appropriate siRNA sequenceit is possible to restrain the target gene expression whichcauses diseases Anderson and coworkers have successfullydeveloped a new siRNA delivery system by incorporatingsix double-stranded siRNAs to tetrahedral DNA assembliesThe single-stranded overhangs on DNA strands allow thespecific hybridization of complementary siRNA sequencesand cancer targeting ligands with better control over theirspatial orientation locations and density These nanostruc-tures have been applied in female BALBc nude mice modelbearing Luc-KB tumorThey found that RNA-modified DNAnanostructures are able to knock down the luciferase levelsin terms of the protein and mRNA levels leading to targetgenes silencing in tumor cells Importantly they exhibit alonger blood circulation time than the parent siRNAs doThiswork highlights the significance of DNA nanostructures toimprove the biostability of tethered RNA strand thus greatlyenhancing the RNAi efficacy in nanomedicine [55]

Recently Sleimanrsquos group has integrated the fireflyLuciferase antisense strands into the DNA triangular prism

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 8: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 7

FF luciferase-expressing cells

ssPS

Transfection

Transfection

LuminescenceTP4X-PS

Figure 3 A diagram showing the effect on luminescence of bear PS and PS-integrated DNA triangular prism

They demonstrated that DNA prisms composed of antisensestrands can significantly induce gene knockdown in HeLacells without being influenced by conjugating small fluores-cent probes within the structure and by serum conditionsThe RNA-modified DNA prisms maintain gene silencing upto 72 h and are still significantly powerful at an initial stage ofgene knockdown after they are removed (Figure 3) [56]

In addition unmethylated cytosino-phosphate-guanine(CpG) oligonucleotides are classified as therapeutic nucleicacids with a strong immunostimulatory effect [26]The CpGsequences are commonly present in bacterial and naturalviral DNA for immune response invading pathogens in ahost [57 58] Interestingly it is found that CpG oligonu-cleotides can effectively be recognized by endosomal Toll-like receptor 9 (TLR9) and further induce conformationalchanges simultaneously [59 60] This process ultimatelytriggers a signaling cascade which leads to the power-ful immunostimulatory properties of CpG oligonucleotidesThey can be highly used for the immunotherapy of cancerand infectious diseases [61 62] However natural CpGoligonucleotides are easily digested by nucleases in biologicalsystems and difficult to pass through the plasma membraneentering cell and reaching their target sites In this regard it isnecessary to develop a nanocarrier with low cytotoxicity andhigh delivery efficacy for clinical uses of CpG Given that self-assembled well-defined DNA nanostructures are rigid andinsensitive to nuclease digestion several research groups haveappended CpG motifs to multidimensional DNA structuresin order to evaluate their uptake efficiency stability andimmunoregulatory effects

Nishikawa et al designed and assembled aY-shapedDNAunit from three single-stranded DNAs Interestingly CpGsequences have been introduced to these strands [63] Theyfound that Y-shaped DNA units induced a great immuneresponse from RAW2647 cells compared to ss- or ds-DNAsin terms of producing a higher amount of proinflamma-tory cytokines such as tumor necrosis factor-120572 (TNF-120572)and interleukin-6 (IL-6) These units also exhibited higheruptake efficiency in macrophage-like cells than natural dsDNAs Subsequently the same group further applied this Y-shaped DNA unit to assemble dendrite-like nanostructuresSurprisingly they demonstrated even a stronger immune

response by inducing a larger amount of proinflammatorycytokines from RAW2674 cells than the monomer Y-shapedDNA units do [64] Recently Nishikawarsquos group developeda series of nanometer-scale polypodna consisting of CpGmotifs and examined their structural and immunologi-cal properties Particularly for hexa- and octapodna theycould highly induce the secretion of TNF-120572 and IL-6 fromRAW2647 cells Interestingly large numbers of pod couldincrease the cellular uptake but also reduce their stabilityin serum condition This enhanced stimulatory activity sug-gests the importance of the stereochemical property of self-assembled DNA nanostructures

Recently Li and coworkers have successfully devel-oped a DNA tetrahedron as a CpG nanocarrier [43]These nanometer-scale 3D structures are structurally rigidmechanically stable and nontoxicThey are also highly stablein serum condition and resistance to nuclease digestion inlive cultured cells for few hours As compared to ssDNA theCpG-functionalized DNA tetrahedral structures can enterRAW2647 cells efficiently Importantly this tetrahedron actsas a carrier to deliver the CpG therapeutic nucleic acids toacquire immune response The amount of certain cytokinesincluding TNF-120572IL-6 and IL-12 stimulated by them wereremarkably increased than those by ss CpG nucleic acidstrand In addition DNA tetrahedral could load more thanone CpG resulting in even higher stimulatory activity Insuch case the positions of CpG loading can be used tomonitor the dose of drug molecule precisely Additionallyseveral groups have successfully developed a large variety oforigami structures for large amount of CpG loading leadingto a strong immune cell activation in freshly isolated spleencells or in RAW 2647 cells by cytokine production in a highlevel (Figure 4) [46 65] In overall it is highly suggested thatvarious geometries of DNA nanoobjects have shown advan-tages of cellular delivery and immunostimulatory activity ofCpG in macrophage-like cells making DNA nanostructurespromising immunotherapeutic carriers

43 Biotin-Streptavidin Interaction Biotin also called vita-min H is a small molecule and exhibits a strong bindingaffinity to biotin-binding proteins such as avidin or strepta-vidin The high affinity of the biotin-streptavidin interactionnot only offers useful bioanalytical advantages [66]but also

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 9: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

8 Journal of Nanomaterials

DNA-tubes CpG

TLR9Nucleus

Cytokines

CD69

Figure 4 A diagram showing how DNA-tubes CpG go into the cell and functionalize

Staplestrands

M13mp18

Annealing DNA origami

dsDNAdsDNA

intercalatedby

doxorubicin

Cell uptake

Tumor cells Doxorigami

Doxorubicinintercalation

Figure 5 A DNA origami designed for doxorubicin transportation

makes this system to be an attractive model for site-specificloading or positioning of guest molecules in highly orderedDNA assemblies [67 68] Recently Gothelf and coworkershave demonstrated a chemical modification of nucleic acidstrands with biotin allowing for streptavidin binding at pre-cise positions in a well-defined self-assembled DNA origamiscaffold In this study biotin-tethered functional groupsincluding an alkyne an amine and an azide reacted withtheir corresponding reactive groups via either a Huisgen-Sharpless-Medal copper(I) catalyzed click chemistry or N-hydroxysuccinimide chemistry The results of high yieldselective cleavage and bond formation in this study offer thepotential of applying such interaction for site-selective uptakeand triggered release of cargos in a control manner [69]

44 Intercalation In DNA chemistry intercalation is areversible insertion of a guest molecule into double helixof DNA strands The small molecules can interact withnucleobases and disturb the 120587-120587 stacking of between double-stranded DNA (dsDNA) Doxorubicin is one of the mostcommon drugs that can be trapped by DNA nanostructuresIt can intercalate in G-C base pair of DNA strand It is smalland can be trapped by DNA nanomaterials easily [70ndash72]Many newly developed DNA nanocarriers have been testeddue to its simplicity [73 74] There is another example of

doxorubicin carried by DNA origami which can circumventdrug resistance It enters and localizes in resistance humanbreast cancer cell (res-MCF-7) while the free doxorubicincannot enter The DNA origami increases pH of lysosome inresistant cancer cells followed by redistribution of drugThiswould allow them to go to their target site (Figure 5) [73]Zhao and his colleagues have also developed a DNA origamitube for transporting doxorubicin By optimizing the designof nanostructures encapsulation efficiency and the releaserate of the drug can be adjusted [74]

Shen et al and Zhu et al also reported the delivery ofDNA-based structures to cells in the presence of intercalateddyes including SYBR Green and carbazole-based biscyanineas fluorescent cargo [75 76] These dyes can specifically bindto and intercalate with DNA duplex giving out strong fluo-rescence Subsequently the intercalated dyes are completelyreleased and a decrease in fluorescence is observed onceDNAstructures are disrupted by some reasons Importantly theyrealized that the enzymatic degradation of these assemblieslasted for at least few hours in cellular environment resultingin sustainable release of cargo molecules

45 Aptamer-Target Interaction Aptamers are either ssDNAor ssRNA molecule that can selectively bind to certaintargets such as proteins and peptides with high affinity and

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 10: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 9

1AS

2 3

Figure 6 Thrombin binding aptamer is introduced into the design of the DNA origami for tagging thrombin

specificityThesemolecules can be presented in a large varietyof shapes including helices and single-stranded nucleic acidloops due to their intrinsic propensity and versatility todiverse targets They can link to various proteins as wellas other nucleic acids small organic compounds and evenentire organisms [77 78] Yanrsquos Group has demonstrated thefirst example of selective DNA aptamer binding as a powerfulplatform for positioning of proteins in periodic locations ofself-assembled DNA arrays (Figure 6) [79] In these studiesthrombin binding aptamer (TBA) is chosen which is a well-known 15-base nucleic acid aptamer consisting of specificsequence of d(GGTTGGTGTGGTTGG) [80] They foundthat DNA-based array constructed with this TBA can foldinto a unimolecular guanine quadruplex and then selectivelybind to a protein called thrombin with nanomolar affinityThis aptamer-target interactionmechanismwould provide analternative choice for cargo uptake with a larger flexibilityand simplicity Only aptamer sequence is required to beimplemented in the design of DNA nanocarrier

In general aptamers are usually selected from a pool oflarge random sequences Because of their high specificity andease of synthesis they have been widely used for biosensingand diagnostic applications [81] More recently aptamershave become therapeutic candidate as biomedical drugs [8283] Common used human 120572-thrombin aptamer which hastwo binding sites can be readily loaded on self-assembledDNA structures with appropriate design [84 85] Fanrsquos groupdesigned a dynamic DNA tetrahedral nanostructure with ananti-ATP aptamer embedded in one of the edges [60] Thisnanostructure could go into cells and monitor the level ofATP via the ATP-induced aptamer conformational changethat alters the FRET efficiency of a pair of fluorophores (Cy3and Cy5) labeled on the structure

The optical activity of DNA strand used to constructDNA nanomolecules would also affect the structure of nano-materials L-DNA and D-DNA has common structure andliability but once the nanostructure is attached to aptamermismatching in nanocage made by D-DNA may occur L-DNA is a better choice for construction because the structureof cage with aptamer is unchanged [86]

46 DNA-Protein Interaction In a cellular environmentthere are many different kinds of proteins while some ofthem can interact with DNA for various cellular reactionsTranscription factor is one of the examples It has bindingsitewhich can interactwithDNAsequenceKapanidisrsquos group

has demonstrated selective trapping of transcription factor(TF) in DNA cage (Figure 7) [87] Transcription factor is aDNA binding protein which is important in gene regulationTF catabolite activator protein (CAP) is used as cargo inthis experiment The 22 base pair DNA recognition site isintegrated in the DNA tetrahedron With the presence ofcyclic adenosine protein (cAMP) the allosteric effector ofprotein increases the binding affinity of CAP towards thebinding recognition sequence These results suggested thatproteinwould still be trapped inside the cage even it is alreadyformed unlike other passive encapsulation methods TheCAP can be released by degradation of cages in presence ofDNA nuclease I

Liu and his coworkers reported a DNA-based deliverysystem for synthetic vaccines [88] In their design biotiny-lated DNA tetrahedron was used as carrier to deliver antigenstreptavidin (STV) intomicewith the aid of biotin-STV inter-action Interestingly the antigen-modified DNA tetrahedroncomplexes could stimulate strong and continuous antibodyresponses against the antigen in comparison with antigenitself On the other hand unmodified DNA nanostructuresdid not induce any response These results indicated thepromise of the use of self-assembled DNA nanostructuresas a delivery and generic platform for rational design andconstruction of vaccines

47 Encapsulation In addition to specific binding interac-tions between cargos and carriers payloads can also bedirectly loaded into container-like DNA nanostructures viapassive encapsulation Recently Sleimanrsquos group demon-strated the ability of a 3DDNA-based nanoobject to passivelyencapsulate certain sizes of cargos [89] DNA nanotubes oflongitudinal variation structure have been created in whichthey can encapsulate gold nanoparticles of specific sizes toform nanoparticle ldquopea-podrdquo lines It is of note that theldquosievingrdquo ability is very important only specific nanoparticlesizes that match the size of the capsules along the nanotubescould be encapsulated and the process is highly selectiveThis approach allows controlling of the positioning andloading of a wide range of sizes of guest molecules in a preciseway by designing the dimensions of cavities inside the DNAnanoobjects

Sequentially Krishnanrsquos group further applied this strat-egy for the encapsulation of a fluorescent biopolymer forexample FITC-dextran in a synthetic icosahedral DNA-based container Without molecular recognition between

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 11: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

10 Journal of Nanomaterials

Unbound

v1

v4

v3

vvvvvvvvvvv44444v1

v3

v3

v1

v2

v2v2

180∘

Rear

Bound

Front

5nm

Figure 7 The figure is showing that the conformation of bound and unbound CAP integrated in DNA tetrahedron

the host and guest cargomolecules are passively loaded to the3D container during joining the two halves of icosahedron inPBS buffer (Figure 8) [90] They have reported the deliveryof DNA icosahedral encapsulated fluorescent dextran (FD)specifically in cellulo Drosophila hemocytes and in C elegansvia anionic ligand-binding receptor (ALBR) pathway TheFD cargo is a complex branched polysaccharide composedof around 10 kDa 52 nm in sizes It is found that thefunctionality of the encapsulated FITC-FD in living wormsis preserved and the spatially mapping of pH changes duringmaturation of the endosomes in coelomocytes

5 Controlled Releases of Cargo Molecules

To act as a nanocarrier for drug delivery control release ofcargo is another significant issue needed to be consideredcarefully In the following section different approaches willbe explained and discussed in detail

51 A DNA Strand Displacement The cargo trapped inDNA nanotube from Sleimanrsquos group is released by stranddisplacement (Figure 9(a)) [89] The nanotube is partiallyhybridized to one strand and gives some tails Introducing thecompletely complementary DNA to the tails the rigidity ofthe cavity capping gold released Sleiman has demonstratedselective release of cargo molecules in response to a specificexternal DNA strand They have designed and assembled3D DNA nanotubes with encapsulated gold nanoparticle aswell as some modified linking strands consisting of an eight-base overhang protruded from each of their large capsulesAfter a fully complementary eraser DNA strand is added tothese self-assembled nanoobjects the closing linking strandsare erased and hybridized and form a double helix withthe complementary eraser DNA strand The fully doubled-stranded DNA nanotubes become partially single-strandedso that the encapsulated cargos are released simultaneously

This release process is highly selective and fast It is just likeunzipping the clothes As the cavity is more flexible withoutthe rigidified strands the nanogold can be leaked out easily

The same group has also applied the same strand dis-placement technique to release the guest molecules such asthe block copolymer micelles loaded on the RCA-nanotubes(Figure 9(b)) [37] and the Nile red or 16-diphenyl-135-hexatriene (DPH) loaded on dendritic alkyl chains-modifiedDNA cages [91]

Goodman et al has reported the operation of recon-figurable braced 3D DNA nanostructure whose structureswitches precisely and reversibly in response to specificmolecular inputs [92] Four DNA strands are mixed insolution to form a tetrahedron which consists of a hairpinloop on one edge This edge can be expanded by adding afuel DNA strand that is fully complementary to the hairpinregion On the other side the edge can be contracted byadding the eraser DNA strand which displaces the fuel strandvia hybridization of its single-stranded overhang first

52 Addition of Small Molecules To carefully realize thepotential of these 3D DNA nanostructures as nanocarriersthe development of spatiotemporal release of the trappedcargo is of great importance Recently Krishnanrsquos grouphas successfully demonstrated the precise control over theopening of a 3D DNA icosahedron loaded with molecularcargo in response to an external small molecule called cyclic-di-GMP (cdGMP) (Figure 10(a)) [93] Generally speakingcdGMP existed as a second messenger in most bacteria forregulation of various biological processes In their designcdGMP aptamers are chosen and have been introduced tothe icosahedral design Upon binding to cdGMP ligandsthe aptamer undergoes a conformational change by stranddisplacement and then dissociate the polyhedral structuresinto two halves Simultaneously the encapsulated fluorescentdextrans are completely released Therefore we strongly

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 12: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 11

VU5

VL5

Ligatepurify

Figure 8 Cargo molecules are passively loaded onto 3D DNA-based container after joining the two halves of icosahedron in PBS buffer

2times

2times

ES1998400

65

(a)

a

a

+4998400998400

(b)

Figure 9 A DNA nanotube for gold releasing by strand displacement (a) and demonstration of PEG releasing in RCA-DNA nanotube bystrand displacement (b)

envision artificial DNA-based nanostructures as nanotool fordrug loading and targeted delivery because of their ability forselective encapsulation and stimuli-triggered release of cargo

53 pH Adjustment pH adjustment is also a possible stim-ulant for the structural change of DNA nanostructures Thekey element of this structural switching mechanism is i-motif switching It makes use of the properties of Watson-Crick base-pairing and Hoogsteen hydrogen bonding In anacidic environment C is partially protonated as C+ whichcan bind with a G-C nucleobase pairs through Hoogsteen H-bonding in order to generate C+G-C triplets However C+loses one electron and turns back to C under neutral envi-ronment discarding the Hoogsteen H-bonding and C+G-C triplets simultaneously Liu et al reported the first pHresponsive DNA tetrahedron in terms of their reversibleassembly and disassembly in response to solution pH changes(Figure 10(b)) [94] In the current design three-point-starDNA motif can associate with one another to form a DNAtetrahedron in acidic environment (pH at 5) through DNAtriplex formation of cytosine-modified sticky ends Whileunder neutral pH environment the tetrahedron dissociatesinto its building blocks immediately The design can beimproved for drug delivery by adjusting pH value towardsthe formation of DNA tetrahedral We strongly believe that

such pH-responsive behavior in self-assembled DNA nanos-tructures will be important for potential applications suchas controlledtargeted drug release in specific cellular envi-ronments The same group also developed a pH biosensorbased on DNA nanomachine which is triggered by protonsto map temporal and spatial pH changes in a cellular systemvia similar structural switching mechanism [95]

54 Photo Irradiation Compared with the above input sig-nals photon is an ideal external source for precise controlof photo-manipulation of DNA nanoobject By using lightDNA nanoobjects can be remotely controlled offering anovel avenue in nanomedicine and drug delivery Generallyspeaking photo irradiation is a clean switching mechanismNO waste is generated as only light was used to drive theentire process It offers capability to precise control lightirradiation in both temporal and spatial fashions Moreimportantly it would not damage the samples as photoirradiation is noninvasive and noncontact source of stimulusRecently azobenzene has been confirmed to be a photo-responsive molecule that can be conjugated to nucleic acidstrands for the regulation of hybridization-dehybridizationprocess [96 97] It exhibited reversible stereoisomerizationproperty It switches from the trans to cis conformation whenexcited at 330ndash380 nmwavelength of light On the other sideit reversibly switches from cis to trans under excitation of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 13: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

12 Journal of Nanomaterials

59984005998400

3998400

5998400

3998400

39984005998400

3998400

times5 times5

cdGMPaddition

FD10encapsulation FD10el

Controlledrelease+ VL5

VUapt5

(a)

pH 50

pH 80

(b)

Figure 10 (a) By binding the cdGMP to aptamer integrated in DNA icosahedron nanocage can be opened for molecule releasing (b) Itmakes use of theWatson-Crick base pair and Hoogsteen base pair properties to construct a DNA tetrahedron which can form in low pH anddecompose in high pH conditions

light with wavelength above 400 nm This intrinsic propertyof azobenzene allows the photo-manipulation ofDNAnanos-tructures in a precise and control manner On the basis of thistechnique Liang et al designed photon-fuelled molecularDNA tweezers consisted of photoresponsive azobenzene-modified DNA strand Photo-induced opening and closingof the tweezers is governed by the irradiation wavelength(Figure 11(a)) [98] Subsequently the same group has success-fully designed and constructed a supra-photoswitch consist-ing of alternating natural nucleobase pairs and azobenzenemoieties in the form of (AAB)n where A and B representthe natural nucleotides and the azobenzene respectively [99]They found that the stability of the azobenzene modifiedDNA duplex is more stable than the neutral one This prop-erty is useful in implementing in different DNAnanocarriersKang et al designed and constructed photoswitchable single-molecular DNA motor with tethered azobenzene moiety[100]This nanomotor is driven by photo irradiation betweenUV light and visible light without any additional DNA strandas external fuel

Recently highly complex DNA nanostructures incorpo-rated with photo-responsive molecule have been successfullydesigned and generated Zou and his coworkers constructedDNA nanoscissors composed of two hairpin structures H1and H2 In this study a DNAzyme is used as an examplesystem for DNA cleavage (Figure 11(b)) [101] Particularly H2is a complementary azobenzene-functionalized sequence atthe 5-end of DNAzyme Under visible light irradiation thetwo hairpins preserve their hairpin structures as duplexesblocking the substrate binding and closing down DNAcleavage activityThis is in a closed state ofDNAnanoscissorsWhile under UV light irradiation H2 is able to be openeddue to structural isomerization of azobenzene from its planarto nonplanar conformations prohibiting duplex formation atH2 and then allowing intermolecular hybridization betweenDNAzyme and the substrate thus activating the enzymatic

activity This is in an open state of DNA nanoscissors Theyfound that the ON and OFF states of nanoscissors lead toa remarkable change in substrate binding affinity and anobvious difference in the activity of DNA cleavage

Yang and his colleagues have successfully demonstratedthe reversible assembly and disassembly of DNA-based struc-tures by introducing azobenzene-modified DNA strands intohexagonalDNAorigami units [102] Anumber of nanometer-sized hexagonal DNA origami structures functionalized withphoto-responsive oligonucleotides have been generatedTheycan be assembled into a large variety of 2D regular orirregular nanostructures under visible irradiation On theother hand DNA hexagonal origami would obtain the cis-conformation under UV light irradiation such that theycannot hybridize together due to steric hindrance effects Byaltering the numbers and positions of azobenzene-modifiedoligonucleotides in the hexagonal shaped DNA origamiscaffolds they can link together in multiorientations in orderto achieve different patterns and configurations criticallyThisphoto irradiation switchingmechanism shows great potentialfor the applications in bionanotechnology such as remote andcontrollable drug release

Based on the above studies we strongly believed thatphoto-triggered release of drug molecules frommultidimen-sional DNA-based nanocarriers would become a promisingrelease mechanism and be highly achievable by carefuldesigns In an advance study Han and coworkers havesuccessfully introduced azobenzene moieties into 3D DNAtetrahedron (Figure 11(c)) [103] Strands with introducedazobenzene groups can hybridize with the single-strandedhairpins allowing the control of open and closed state ofDNA tetrahedron by visible and UV light The hybridizationand dissociation of azobenzene-modified oligonucleotidescan be remotely and reversibly controlled by the interconver-sion of trans and cis confirmations of azobenzene molecules

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 14: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 13

Closed

Vis

Open

cis

5998400

3998400

HN

N

NN

N

O

P

O

OO OH

UV

trans

(a)

Open DNAnanoscissors

Closed DNAnanoscissors

UV light

Vis light

5998400

3998400

5998400

3998400

HN

N N

O

PO

O

O

OH

NNH

N

O

P

O

O

O OH

(b)

Vis

UV

(c)

Figure 11 (a) A design of photo-sensitive DNA nanodevice that make use of the properties of azobenzene towards different wavelengths oflight (b) Making use of the cis-trans properties of azobenzene under different wavelengths to close or disclose the active site of enzyme (c)The shape of the azobenzene modified DNA tetrahedron can be altered in the presence of different wavelengths

It is believed that these studies will open doors to implementand facilitate the 3D structural changes for triggered-releaseof encapsulated cargos in DNA-based nanoobjects

6 Cellular Internalization and Site-SpecificTargeting of DNA Nanostructures

61 Passive Delivery DNA-based molecules usually havegreat difficulties in delivering to cells as they are highly neg-atively charged They are not able to pass through cell mem-branes directly Most of them undergo three types of possiblemechanisms of getting in cells Clathrin-mediated endocy-tosis Cavolae-mediated endocytosis and macropinocytosisIn general Clathrin-mediated endocytosis is a type of endo-cytois which requires excitation of receptor The moleculeswould then be trapped in early endosome then in lateendosome and finally in lysosome The pH in a cellularenvironment is gradually decreased and then degradationof self-assembled DNA nanostructures is highly possibleCaveolae-mediated endocytosis is another type of endocyto-sis but it would go to Caveosome and then migrate to Golgiendoplasmic reticulum and endosomes Macropinocytosisis different from the above two endocytic pathways as it isnonspecificThough themolecules should end up at lysosomebut the macropinisome is comparatively leaky which make

them possible to enter the cytosol to escape the destiny ofdegradation [104ndash106] Efforts have been put to improve thecellular uptake of DNA-based nanomaterials in terms of highcell penetration ability and low cytotoxicity [107 108]

62 Targeting of Self-Assembled DNA Nanostructures Toenhance the selective delivery of DNA nanocarriers to cancercells or particular intracellular organelles for drug deliverypurposes a targeting moiety has to be conjugated to DNAassemblies

621 Folate Folate water-soluble vitamin B9 has proven

to be an efficient targeting agent for cancer cells as folatereceptors are overexpressed on the surfaces of cancer cellsTherefore DNA nanostructures decorated with folate groupvia a simple NHS chemistry would provide a higher chanceto be taken up by cancer cells over normal cells Maorsquos groupintegrates folate into his DNA nanotubes (Figure 12(a)) [51]They prove that the folate modified DNA nanotubes enterKB cells through overexpressed folate receptor and be able tointernalize in the cellsOnehour incubation of thesemodifiednanotubes would be saturated because cells may only be ableto take up certain amount of DNA nanotubes When thefolate content in the DNA nanostructures reaches 10 theuptake capability of DNA nanotubes in cells would reachplateau due to the limited number of folate receptors

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 15: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

14 Journal of Nanomaterials

Cancer cellFolate receptor (FR)

Dual-functionalizedDNA nanotubes (NT)

Cy3

Single strandedDNA (ssDNA)

Folate

(a)

DoxoApt-DNA-icosa

DoxoApt-DNA-icosa

MUC1

Earlyendosome

Lateendosome

Doxo

Doxo

Doxo

Cytosol

Doxo

Doxo

Doxo

DoxoDoxo Doxo

Doxo

Doxo

DoxoDoxo

Doxo

Nucleus

Lysosome

Six-point-star motif

Apt-DNA-icosaDoxorubicin

Doxo

erectedAptamer

pH darr

pH darr

pH darr

(b)

Figure 12 (a) Cy3 and folate is covalently conjugated to the ssDNA via NHS chemistry for cell targeting and visualization (b) The figure isshowing the design of the DNA icosahedral nanoparticles and the possible releasing mechanism of doxorubicin

622 Aptamer In general aptamers are short single-stranded nucleic acid strands with specific sequences derivedfrom systematic evolution of ligands by exponential enrich-ment (SELEX)They are able to recognize and bind to cellularsurface receptors in certain cancer cells and thus allowimporting to the cells leading to target delivery Huangrsquosgroup have designed a DNA icosahedra from a six-point-starmotif with a sticky end segment of MUC 1 aptamer sequence(Figure 12(b)) [109] MUC 1 is a major class of tumorsurface marker which is abundant on the surface of mostepithelial cancer cells [110 111] serving as entering portalsfor aptamers [112] To investigate the targeting selectivity the

uptakes of DNA polyhedron byMCF-7 cells which areMUC-receptor positive tumor cells and by CHO-K1 cells whichare MUC-receptor negative cells have been investigatedThey found that aptamer-modifiedDNApolyhedra exhibitedhigher cellular internalization efficiency than the regularDNA polyhedra do in MCF-7 cells but not in CHO-K1 cellsconfirming an aptamer-mediated cellular selectivity of inter-nalization of DNA polyhedra They have proposed a cellularuptake mechanism for aptamer-modified DNA polyhedra inMCF-7 cells FirstMUC-modifiedDNApolyhedra recognizeMUC 1 which is then rapidly recycled through intracellularcompartments After that MUC-modified DNA polyhedral

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 16: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 15

Functional domains Connector

Cell uptake

Arms Aptamer MDR1-ASNH

OAcrydite

AptNAs

h

Building unit

Self-assembly

(a)

Weak emission state

Staple strands

annealing

M13 genome DNA Tubular DNA origami

Free probes

incubation

Origami-probe complexStrong emission

N+

IminusIminus N+

NC5H11

(b)

Figure 13 (a) DNA strand are modified to bind with different functional domains and photosynthesized to a bigger complex Thenanostructure contains aptamer for differential cell targeting (b) A design of label-free fluorescent probe incorporated in DNA origami

structures are smuggled to endosome and later to lysosomeby binding to MUC 1

Tanrsquos group have successfully designed and generatedmultifunctional DNA nanoassembly by first self-assemblingthree components including aptamer acrydite-modifiedssDNA and antisense oligonucleotides to form Y-shapedDNA domains (Figure 13(a)) [70] Subsequently these func-tional DNA domains were hybridized to an X-shaped DNAconnector to form building units After photo irradiation allbuilding units were cross-linked to form aptamer-basedDNAassemblies In this study sgc8 aptamer and KK1B10 aptamerwere chosen to demonstrate the generality of selective recog-nition of target cancer cells by thesemultifunctional aptamer-based nanoassemblies Their results indicated that sgc8-functionalized DNA assemblies internalized specifically toCCRF-CEMcancer cells (T cell acute lymphoblastic leukemiacell line) but not to Ramos cells (B cells human Burkittrsquoslymphoma) While KK1B10 can specifically recognize andinternalize into K562D (Dox-resistant leukemia cell line)

but cannot control Ramos cells Using this technique theconstruction of the nanocarrier is easy to achieve and ishighly programmable as the position number and size of theaptamer can be adjusted In addition this system has beentested in vitro indicating that the nanoassembly is enzymaticresistant and cytotoxic negligible

Recently Kim et al decorated their l-DNA nanocarrierswith antiproliferative aptamer AS1411 allowing them toselectively recognize and take up by cancer cells [86] This islikely due to the interaction between AS1411 aptamers on l-DNA nanocarriers and the target protein nucleolin expressedon the surface of HeLa cells

623 Organelle Localization Signal Peptides Most of the self-assembled DNA nanostructures are taken up and eventuallylocalized in lysosomes endosomes or Golgi networks bymeans of endocytosis (Figure 13(b)) [75 95 113] It is realizedthat these locations are highly limited by their biological

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 17: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

16 Journal of Nanomaterials

30min

APTMS coated SiNNs

DNA nanocagesuspension

HeLa cell

4h

Remove cell from SiNWsand replate on coverslip

NH3

NH3 NH3

NH3

OOOO

OO

O

OOO

OO

Si

SiSi

Si

(a)

Single-strandedCy5-labeled DNA-NC

Peptide-functionalized RS

Peptide-functionalizedCy5-labeled DNA-NC

DNA oligos 3998400-AATAATTTCAGAGTCTTTTTT-5998400HN-peptidesMTS HN-120573ALLYRSSCLTRTAPKFFRISQRLSLMNTS HN-120573AVVVKKKRKVVC

(b) (c)

(d)

Figure 14 (a) Demonstration of how functionalized vertical silicon nanowire arrays help in direct delivery of molecules to cytosol (b) Thetriangular prism has been attached to MTS and NTS for specific cell internal targeting to mitochondria and nucleus respectively (c) Cy5-labeled MTS DNA-NCs with MitoTracker green and Cy5-labeled (Scale bar represents 15 120583m)

behaviors and functions in a cellular system among differentintracellular compartments

Our group recently developed a new delivery technologyon the basis of functionalized vertical silicon nanowire arraysas a delivery platform to transport intact DNA cages to thecytosol efficiently without endocytosis (Figure 14) [52] Weproved that this delivery strategy exhibits high cellular uptakeefficiency together with great stability and low cytotoxicityin a cellular environment In addition this delivery approachwould preserve the structural integrity of cages and help themescape degradation under endocytosis More importantlywe demonstrated the first example of site-selective DNAnanocages for targeting mitochondria and nuclei In thisstudy specific organelle localization signal peptides such asmitochondrial localization signal (MLS) peptide or nucleus

localization signal (NLS) peptide were incorporated to oneof the constituent DNA strands and then further assembledto MLS or NLS peptide-functionalized DNA nanocage Itis found that the modified MLS or NLS-cages are able tolocalize exclusively inmitochondria or nuclei respectively bymeans of a powerful SiNW delivery platform in vitro Thiswork opens a door for the use of DNA nanocage as smartvehicles particularly for targeted drug delivery to the specificintracellular organelles

7 Conclusions and Outlook

DNA nanotechnology becomes a cutting edge research inrecent yearsThe role of DNA in nanotechnology has reachedfar beyond its intrinsic role in biology With the well-known

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 18: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 17

knowledge of self-recognition properties of DNAand its dou-ble helix feature on the molecular level different geometriesand sizes of DNA-based nanoarchitectures can be generatedvery accurately and efficiently in contrast to other self-assembling systems In this review article we summarizedrecent progress of drug delivery system based on multidi-mensional DNA nanostructures Thus self-assembled DNAnanostructures are undoubtedly highly promising scaffoldto act as a drug nanocarrier or to display functionalitiesfor therapeutic applications From the high demand ofmultifunctional DNA carriers in the context of drug deliveryvehicles that have been described in detail here we cansummarize several reasons why self-assembled nucleic acidstructures are feasible for targeted drug delivery First theDNA nanostructures can be designed and modified withmultifunctional groups including drug molecules targetingmotifs and fluorescence probes and position all of themwith high accuracy Second in comparison with multistepsynthesis of other nanocarrier scaffolds like dendrimers thedesired DNA nanoobjects with great versatility can be easilyformed by simple mixing of individual DNA building blockstogether in a single stepThis strategy can be achieved a largesize of DNA nanostructures effortlessly ranging from only afew nanometers to micrometer scale Another conspicuousfeature suggesting the use of self-assembled DNA-basedcarrier is that they can pass through the negatively chargedplasma membrane and get into the cells efficiently withoutthe need of transfecting agents except some of the largeand flexible DNA origami structures as compared to nakedDNA strand itself In addition all DNA-based nanomaterialsexhibited a very low cytotoxicity no matter in the presenceor absence of the payloads or stimuli Such feature makes theself-assembled DNA nanoarchitectures a promising deliverysystem Another striking advantage of using DNA nanoob-jects for the purpose of drug delivery is that a large number ofdrug loading methods have been utilized for the interactionbetween drug moleculescargos and self-assembled DNAnanostructures We have described the examples briefly inthis paper They included covalent linkage nucleic acidbase-pairing biotin-streptavidin interaction intercalationaptamer-targeted interaction DNA-protein interaction andencapsulation Scientists also demonstrated several possi-bilities for the control release of drug or cargo moleculesIn the presence of the specific and weak hydrogen bondsbetween A and T and C and G nucleobases a stranddisplacement is a method by adding an eraser DNA or RNAstrand allowing exchange and release of strands consistingof a toehold overhang This DNA-mediated release strategyhighly relies on specific nucleic acid sequences When thoseDNA nanostructures are introduced into an environmentwith different pH values i-motif switching is a promisingmechanism for structural change and control release of cargosimultaneously Another option for drug release is the useof light In this case light acts as a stimulus to facilitate theclean removal process No accumulation of waste happensOverall multifunctional DNA nanostructures have success-fully demonstrated their efficient intracellular delivery andspecific targeting to cancer cells or particular intracellularorganelles including lysosomes endosomes Golgi networks

mitochondria and the nuclei They are also extensively usedfor the delivery of certain drug or cargo molecules in livingcell systems and induced some cellular activities or effectsaccordingly To sum up self-assembled DNA nanostructuresoffer unprecedented control over their structures and func-tionalities in a biological or cellular environment the aboveexamples demonstrate the potential applications particularlyfor targeted drug delivery or gene regulation

However the use of DNA nanostructures in the biomed-ical field faces several challenges As self-assembled DNAnanostructures have been seriously considered for the appli-cation in drug delivery further studies are needed to obtainbetter information for their practical applicationsThese con-sist of the understanding of cellular uptake mechanism suchas their intracellular pathway and pharmacokinetics Canthey escape from the fate of being degraded by endocytosisbefore reaching the target sites and taking biological effectsIt is also necessary to investigate the relationship betweentheir intracellular behaviorfunction and their various chem-icalphysical properties such as functional group incorpora-tion surface charges nucleobase sequences geometry anddimensions Another focus which should be concentratedon is the study of selective targeting of functionalized DNAnanostructures in terms of discrimination of diseased cellsfrom common normal cells in vitro and in vivo For instancehow can they be only taken up by cancer cells but notmacrophages It is also important to look for some chemicalmodifications to prevent the formation of aggregates incirculating systemandovercome themultilayers barriers afterthe DNA-based nanocarriers enter human body The lastbut not the least an alternative new and safe control releasemechanism for drugmolecules should be developed such thatno waste is accumulated in biological system in addition tono harm being induced to the tissues of human bodies Westrongly believe that these suggested questions and studies areattractive topics to be investigated in the near future

Conflict of Interests

The authors declare that there is no conflict of interestsregarding the publication of this paper

Acknowledgments

This work was supported by National Science Foundationof China 21324077 CityU Strategic Research Grant 7004026and CityU Start-up Grant 7200300

References

[1] J AHubbell andA Chilkoti ldquoNanomaterials for drug deliveryrdquoScience vol 337 no 6092 pp 303ndash305 2012

[2] M H El-Dakdouki E Pure and X Huang ldquoDevelopment ofdrug loaded nanoparticles for tumor targeting Part 1 synthesischaracterization and biological evaluation in 2D cell culturesrdquoNanoscale vol 5 no 9 pp 3895ndash3903 2013

[3] L Moore E K-H Chow E Osawa J M Bishop and D HoldquoDiamond-lipid hybrids enhance chemotherapeutic tolerance

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 19: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

18 Journal of Nanomaterials

and mediate tumor regressionrdquo AdvancedMaterials vol 25 no26 pp 3532ndash3541 2013

[4] K Salazar-Salinas CKubli-Garfias and JM Seminario ldquoCom-putational design of a CNT carrier for a high affinity bispecificanti-HER2 antibody based on trastuzumab and pertuzumabFabsrdquo Journal of Molecular Modeling vol 19 no 7 pp 2797ndash2810 2013

[5] M E Davis J E Zuckerman C H J Choi et al ldquoEvidenceof RNAi in humans from systemically administered siRNA viatargeted nanoparticlesrdquo Nature vol 464 no 7291 pp 1067ndash1070 2010

[6] K Miyata R J Christie and K Kataoka ldquoPolymeric micellesfor nano-scale drug deliveryrdquoReactive and Functional Polymersvol 71 no 3 pp 227ndash234 2011

[7] Y L Colson and M W Grinstaff ldquoBiologically responsivepolymeric nanoparticles for drug deliveryrdquoAdvancedMaterialsvol 24 no 28 pp 3878ndash3886 2012

[8] H Pei X Zuo D Zhu Q Huang and C Fan ldquoFunctionalDNA nanostructures for theranostic applicationsrdquo Accounts ofChemical Research vol 47 no 2 pp 550ndash559 2014

[9] C Song Z-GWang and B Ding ldquoSmart nanomachines basedonDNAself-assemblyrdquo Small vol 9 no 14 pp 2382ndash2392 2013

[10] L M Smith ldquoNanostructures the manifold faces of DNArdquoNature vol 440 no 7082 pp 283ndash284 2006

[11] J Fu M Liu Y Liu and H Yan ldquoSpatially-interactivebiomolecular networks organized by nucleic acid nanostruc-turesrdquo Accounts of Chemical Research vol 45 no 8 pp 1215ndash1226 2012

[12] N C Seeman ldquoNucleic acid junctions and latticesrdquo Journal ofTheoretical Biology vol 99 no 2 pp 237ndash247 1982

[13] N C Seeman ldquoDNA in a material worldrdquo Nature vol 421 no6921 pp 427ndash431 2003

[14] N R Kallenbach R I Ma and N C Seeman ldquoAn immo-bile nucleic acid junction constructed from oligonucleotidesrdquoNature vol 305 no 5937 pp 829ndash831 1983

[15] R-I Ma N R Kallenbach R D Sheardy M L Petrillo andN C Seeman ldquoThree-arm nucleic acid junctions are flexiblerdquoNucleic Acids Research vol 14 no 24 pp 9745ndash9753 1986

[16] E Winfree F Liu L A Wenzler and N C Seeman ldquoDesignand self-assembly of two-dimensional DNA crystalsrdquo Naturevol 394 no 6693 pp 539ndash544 1998

[17] H Yan S H Park G Finkelstein J H Reif and T H LaBeanldquoDNA-templated self-assembly of protein arrays and highlyconductive nanowiresrdquo Science vol 301 no 5641 pp 1882ndash1884 2003

[18] D Liu M Wang Z Deng R Walulu and C Mao ldquoTensegrityconstruction of rigid DNA triangles with flexible four-armDNA junctionsrdquo Journal of the American Chemical Society vol126 no 8 pp 2324ndash2325 2004

[19] CMaoW Sun andN C Seeman ldquoDesigned two-dimensionalDNA holliday junction arrays visualized by atomic forcemicroscopyrdquo Journal of the American Chemical Society vol 121no 23 pp 5437ndash5443 1999

[20] C Zhang Y He M Su et al ldquoDNA self-assembly from 2D to3Drdquo Faraday Discussions vol 143 pp 221ndash233 2009

[21] C Lin Y Liu and H Yan ldquoDesigner DNA nanoarchitecturesrdquoBiochemistry vol 48 no 8 pp 1663ndash1674 2009

[22] Z-G Wang and B Ding ldquoDNA-based self-assembly for func-tional nanomaterialsrdquo Advanced Materials vol 25 no 28 pp3905ndash3914 2013

[23] P W K Rothemund ldquoFolding DNA to create nanoscale shapesand patternsrdquo Nature vol 440 no 7082 pp 297ndash302 2006

[24] H Yan T H LaBean L Feng and J H Reif ldquoDirected nucle-ation assembly of DNA tile complexes for barcode-patternedlatticesrdquo Proceedings of the National Academy of Sciences of theUnited States of America vol 100 no 14 pp 8103ndash8108 2003

[25] W M Shih J D Quispe and G F Joyce ldquoA 17-kilobase single-stranded DNA that folds into a nanoscale octahedronrdquo Naturevol 427 no 6975 pp 618ndash621 2004

[26] SM Douglas H Dietz T Liedl B Hogberg F Graf andWMShih ldquoSelf-assembly of DNA into nanoscale three-dimensionalshapesrdquo Nature vol 459 no 7245 pp 414ndash418 2009

[27] Z Li M Liu L Wang J Nangreave H Yan and Y LiuldquoMolecular behavior of DNA origami in higher-order self-assemblyrdquo Journal of the AmericanChemical Society vol 132 no38 pp 13545ndash13552 2010

[28] Z-G Wang C Song and B Ding ldquoFunctional DNA nanos-tructures for photonic and biomedical applicationsrdquo Small vol9 no 13 pp 2210ndash2222 2013

[29] V Linko andH Dietz ldquoThe enabled state of DNA nanotechnol-ogyrdquo Current Opinion in Biotechnology vol 24 no 4 pp 555ndash561 2013

[30] A-P Eskelinen H Rosilo A Kuzyk P Torma and M A Kos-tiainen ldquoControlling the formation of DNA origami structureswith external signalsrdquo Small vol 8 no 13 pp 2016ndash2020 2012

[31] C K McLaughlin G D Hamblin and H F SleimanldquoSupramolecularDNAassemblyrdquoChemical Society Reviews vol40 no 12 pp 5647ndash5656 2011

[32] F A Aldaye P K Lo P Karam C K McLaughlin G Cosaand H F Sleiman ldquoModular construction of DNA nanotubesof tunable geometry and single- or double-stranded characterrdquoNature Nanotechnology vol 4 no 6 pp 349ndash352 2009

[33] P K Lo F Altvater and H F Sleiman ldquoTemplated synthesisof DNA nanotubes with controlled predetermined lengthsrdquoJournal of the American Chemical Society vol 132 no 30 pp10212ndash10214 2010

[34] H Yang F Altvater A D de Bruijn C K McLaughlin P K Loand H F Sleiman ldquoChiral metal-DNA four-arm junctions andmetalated nanotubular structuresrdquoAngewandte Chemie vol 50no 20 pp 4620ndash4623 2011

[35] Y Wen C K McLaughlin P K Lo H Yang and H FSleiman ldquoStable gold nanoparticle conjugation to internal DNApositions facile generation of discrete gold nanoparticle-DNAassembliesrdquoBioconjugate Chemistry vol 21 no 8 pp 1413ndash14162010

[36] H Yang K L Metera and H F Sleiman ldquoDNA modified withmetal complexes applications in the construction of higherorder metal-DNA nanostructuresrdquo Coordination ChemistryReviews vol 254 no 19-20 pp 2403ndash2415 2010

[37] K M M Carneiro G D Hamblin K D Hanni et alldquoStimuli-responsive organization of block copolymers on DNAnanotubesrdquo Chemical Science vol 3 no 6 pp 1980ndash1986 2012

[38] P K Lo K L Metera and H F Sleiman ldquoSelf-assembly ofthree-dimensional DNA nanostructures and potential biolog-ical applicationsrdquo Current Opinion in Chemical Biology vol 14no 5 pp 597ndash607 2010

[39] J Li C Fan H Pei J Shi and Q Huang ldquoSmart drug deliv-ery nanocarriers with self-assembled DNA nanostructuresrdquoAdvanced Materials vol 25 no 32 pp 4386ndash4396 2013

[40] E S AndersenM DongMM Nielsen et al ldquoSelf-assembly ofa nanoscale DNA box with a controllable lidrdquo Nature vol 459no 7243 pp 73ndash76 2009

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 20: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 19

[41] J-W Keum and H Bermudez ldquoEnhanced resistance of DNAnanostructures to enzymatic digestionrdquo Chemical Communica-tions no 45 pp 7036ndash7038 2009

[42] A S Walsh H Yin C M Erben M J A Wood and AJ Turberfield ldquoDNA cage delivery to mammalian cellsrdquo ACSNano vol 5 no 7 pp 5427ndash5432 2011

[43] J Li H Pei B Zhu et al ldquoSelf-assembled multivalentDNA nanostructures for noninvasive intracellular delivery ofimmunostimulatory CpG oligonucleotidesrdquo ACS Nano vol 5no 11 pp 8783ndash8789 2011

[44] QMei XWei F Su et al ldquoStability ofDNAorigami nanoarraysin cell lysaterdquo Nano Letters vol 11 no 4 pp 1477ndash1482 2011

[45] C E Castro F Kilchherr D-N Kim et al ldquoA primer toscaffolded DNA origamirdquoNatureMethods vol 8 no 3 pp 221ndash229 2011

[46] V J Schuller S Heidegger N Sandholzer et al ldquoCellularimmunostimulation by CpG-sequence-coated DNA origamistructuresrdquo ACS Nano vol 5 no 12 pp 9696ndash9702 2011

[47] J W Conway C K McLaughlin K J Castor and H SleimanldquoDNAnanostructure serum stability greater than the sum of itspartsrdquo Chemical Communications vol 49 no 12 pp 1172ndash11742013

[48] G D Hamblin K M M Carneiro J F Fakhoury K E BujoldandH F Sleiman ldquoRolling circle amplification-templated DNAnanotubes show increased stability and cell penetration abilityrdquoJournal of the American Chemical Society vol 134 no 6 pp2888ndash2891 2012

[49] K E Bujold J Fakhoury T G W Edwardson et al ldquoSequence-responsive unzipping DNA cubes with tunable cellular uptakeprofilesrdquo Chemical Science vol 5 no 6 pp 2449ndash2455 2014

[50] C K McLaughlin G D Hamblin K D Hanni et al ldquoThree-dimensional organization of block copolymers on ldquoDNA- min-imalrdquo scaffoldsrdquo Journal of the American Chemical Society vol134 no 9 pp 4280ndash4286 2012

[51] S Ko H Liu Y Chen and C Mao ldquoDNA nanotubes ascombinatorial vehicles for cellular deliveryrdquoBiomacromoleculesvol 9 no 11 pp 3039ndash3043 2008

[52] M S Chan and P K Lo ldquoNanoneedle-assisted delivery of site-selective peptide-functionalized DNA nanocages for targetingmitochondria and nucleirdquo Small vol 10 no 7 pp 1255ndash12602014

[53] S M Douglas I Bachelet and G M Church ldquoA logic-gated nanorobot for targeted transport of molecular payloadsrdquoScience vol 335 no 6070 pp 831ndash834 2012

[54] C Zhang X Li C Tian et al ldquoDNA nanocages swallow goldnanoparticles (AuNPs) to form AuNPDNA cage core-shellstructuresrdquo ACS Nano vol 8 no 2 pp 1130ndash1135 2014

[55] H Lee A K R Lytton-Jean Y Chen et al ldquoMolecularly self-assembled nucleic acid nanoparticles for targeted in vivo siRNAdeliveryrdquoNatureNanotechnology vol 7 no 6 pp 389ndash393 2012

[56] J J Fakhoury C K McLaughlin T W Edwardson J WConway andH F Sleiman ldquoDevelopment and characterizationof gene silencing DNA cagesrdquo Biomacromolecules vol 15 no 1pp 276ndash282 2014

[57] A Bianco J Hoebeke S Godefroy et al ldquoCationic carbonnanotubes bind to CpG oligodeoxynucleotides and enhancetheir immunostimulatory propertiesrdquo Journal of the AmericanChemical Society vol 127 no 1 pp 58ndash59 2005

[58] AM Krieg ldquoImmune effects andmechanisms of action of CpGmotifsrdquo Vaccine vol 19 no 6 pp 618ndash622 2000

[59] H Hemmi O Takeuchi T Kawai et al ldquoA Toll-like receptorrecognizes bacterial DNArdquo Nature vol 408 no 6813 pp 740ndash745 2000

[60] M Heikenwalder M Polymenidou T Junt et al ldquoLymphoidfollicle destruction and immunosuppression after repeatedCpGoligodeoxynucleotide administrationrdquoNature Medicine vol 10no 2 pp 187ndash192 2004

[61] M Schmidt K Anton C Nordhaus C Junghans B Wittigand MWorm ldquoCytokine and Ig-production by CG-containingsequences with phosphorodiester backbone and dumbbell-shaperdquo Allergy vol 61 no 1 pp 56ndash63 2006

[62] M Wei N Chen J Li et al ldquoPolyvalent immunostimu-latory nanoagents with self-assembled CpG oligonucleotide-conjugated gold nanoparticlesrdquoAngewandte Chemie vol 51 no5 pp 1202ndash1206 2012

[63] M Nishikawa M Matono S Rattanakiat N Matsuokaand Y Takakura ldquoEnhanced immunostimulatory activity ofoligodeoxynucleotides byY-shape formationrdquo Immunology vol124 no 2 pp 247ndash255 2008

[64] S Rattanakiat M Nishikawa H Funabashi D Luo and YTakakura ldquoThe assembly of a short linear natural cytosine-phosphate-guanine DNA into dendritic structures and its effecton immunostimulatory activityrdquo Biomaterials vol 30 no 29pp 5701ndash5706 2009

[65] X Ouyang J Li H Liu et al ldquoRolling circle amplification-based DNA origami nanostructrures for intracellular deliveryof immunostimulatory drugsrdquo Small vol 9 no 18 pp 3082ndash3087 2013

[66] M Wilchek and E A Bayer ldquoThe avidin-biotin complex inbioanalytical applicationsrdquo Analytical Biochemistry vol 171 no1 pp 1ndash32 1988

[67] P C Weber M W Pantoliano and L D Thompson ldquoCrystalstructure and ligand-binding studies of a screened peptidecomplexed with streptavidinrdquo Biochemistry vol 31 no 39 pp9350ndash9354 1992

[68] WAHendricksonA Pahler J L Smith Y Satow E AMerrittand R P Phizackerley ldquoCrystal structure of core streptavidindetermined from multiwavelength anomalous diffraction ofsynchrotron radiationrdquo Proceedings of the National Academy ofSciences of the United States of America vol 86 no 7 pp 2190ndash2194 1989

[69] N V Voigt T Toslashrring A Rotaru et al ldquoSingle-moleculechemical reactions on DNA origamirdquo Nature Nanotechnologyvol 5 no 3 pp 200ndash203 2010

[70] C Wu D Han T Chen et al ldquoBuilding a multifunctionalaptamer-based dna nanoassembly for targeted cancer therapyrdquoJournal of the American Chemical Society vol 135 no 49 pp18644ndash18650 2013

[71] MHuybrechtsM Symann andA Trouet ldquoEffects of daunoru-bicin and doxorubicin free and associated with DNA onhemopoietic stem cellsrdquo Cancer Research vol 39 no 9 pp3738ndash3743 1979

[72] A Bodley L F Liu M Israel et al ldquoDNA topoisomerase II-mediated interaction of doxorubicin and daunorubicin con-geners with DNArdquo Cancer Research vol 49 no 21 pp 5969ndash5978 1989

[73] Q Jiang C Song J Nangreave et al ldquoDNA origami as a carrierfor circumvention of drug resistancerdquo Journal of the AmericanChemical Society vol 134 no 32 pp 13396ndash13403 2012

[74] Y-X Zhao A Shaw X Zeng E Benson A M Nystrom andB Hogberg ldquoDNA origami delivery system for cancer therapy

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 21: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

20 Journal of Nanomaterials

with tunable release propertiesrdquo ACS Nano vol 6 no 10 pp8684ndash8691 2012

[75] X Shen Q Jiang J Wang et al ldquoVisualization of the intracel-lular location and stability of DNA origami with a label-freefluorescent proberdquo Chemical Communications vol 48 no 92pp 11301ndash11303 2012

[76] G Zhu S Zhang E Song et al ldquoBuilding fluorescent DNAnanodevices on target living cell surfacesrdquoAngewandte Chemievol 52 no 21 pp 5490ndash5496 2013

[77] E N Brody M C Willis J D Smith S Jayasena D Zichiand L Gold ldquoThe use of aptamers in large arrays for moleculardiagnosticsrdquo Molecular Diagnosis vol 4 no 4 pp 381ndash3881999

[78] J C Cox and A D Ellington ldquoAutomated selection of anti-protein aptamersrdquo Bioorganic and Medicinal Chemistry vol 9no 10 pp 2525ndash2531 2001

[79] Y Liu C Lin H Li and H Yan ldquoAptamer-directed self-assembly of protein arrays on a DNA nanostructurerdquo Ange-wandte Chemie vol 44 no 28 pp 4333ndash4338 2005

[80] K Padmanabhan K P Padmanabhan J D Ferrara J E Sadlerand A Tulinsky ldquoThe structure of 120572-thrombin inhibited bya 15-mer single-stranded DNA aptamerrdquo Journal of BiologicalChemistry vol 268 no 24 pp 17651ndash17654 1993

[81] S Song L Wang J Li C Fan and J Zhao ldquoAptamer-basedbiosensorsrdquo TrAC Trends in Analytical Chemistry vol 27 no2 pp 108ndash117 2008

[82] V Bagalkot O C Farokhzad R Langer and S Jon ldquoAnaptamer-doxorubicin physical conjugate as a novel targeteddrug-delivery platformrdquo Angewandte Chemie vol 45 no 48pp 8149ndash8152 2006

[83] E Levy-Nissenbaum A F Radovic-Moreno A Z Wang RLanger and O C Farokhzad ldquoNanotechnology and aptamersapplications in drug deliveryrdquo Trends in Biotechnology vol 26no 8 pp 442ndash449 2008

[84] C Zhou Z Yang and D Liu ldquoReversible regulation of proteinbinding affinity by a DNA machinerdquo Journal of the AmericanChemical Society vol 134 no 3 pp 1416ndash1418 2012

[85] W U Dittmer A Reuter and F C Simmel ldquoA DNA-basedmachine that can cyclically bind and release thrombinrdquo Ange-wandte ChemiemdashInternational Edition vol 43 no 27 pp 3550ndash3553 2004

[86] K-R Kim T Lee B-S Kim and D-R Ahn ldquoUtilizing thebioorthogonal base-pairing system of l-DNA to design idealDNAnanocarriers for enhanced delivery of nucleic acid cargosrdquoChemical Science vol 5 no 4 pp 1533ndash1537 2014

[87] R Crawford C M Erben J Periz et al ldquoNon-covalentsingle transcription factor encapsulation inside a DNA cagerdquoAngewandte Chemie vol 52 no 8 pp 2284ndash2288 2013

[88] X Liu Y Xu T Yu et al ldquoA DNA nanostructure platform fordirected assembly of synthetic vaccinesrdquo Nano Letters vol 12no 8 pp 4254ndash4259 2012

[89] P K Lo P Karam F A Aldaye et al ldquoLoading and selectiverelease of cargo in DNA nanotubes with longitudinal variationrdquoNature Chemistry vol 2 no 4 pp 319ndash328 2010

[90] D Bhatia S Surana S Chakraborty S P Koushika andY Krishnan ldquoA synthetic icosahedral DNA-based host-cargocomplex for functional in vivo imagingrdquo Nature Communica-tions vol 2 article 339 2011

[91] T GW Edwardson K MM Carneiro C K McLaughlin C JSerpell andH F Sleiman ldquoSite-specific positioning of dendriticalkyl chains on DNA cages enables their geometry-dependent

self-assemblyrdquo Nature Chemistry vol 5 no 10 pp 868ndash8752013

[92] R P Goodman M Heilemann S Doose C M Erben A NKapanidis andA J Turberfield ldquoReconfigurable braced three-dimensionalDNAnanostructuresrdquoNatureNanotechnology vol3 no 2 pp 93ndash96 2008

[93] A Banerjee D Bhatia A Saminathan S Chakraborty S Karand Y Krishnan ldquoControlled release of encapsulated cargofrom aDNA icosahedron using a chemical triggerrdquoAngewandteChemiemdashInternational Edition vol 52 no 27 pp 6854ndash68572013

[94] Z Liu Y Li C Tian and C Mao ldquoA smart DNA tetrahedronthat isothermally assembles or dissociates in response to thesolution pH value changesrdquo Biomacromolecules vol 14 no 6pp 1711ndash1714 2013

[95] S Modi M G Swetha D Goswami G D Gupta S Mayor andY Krishnan ldquoA DNA nanomachine that maps spatial and tem-poral pH changes inside living cellsrdquo Nature Nanotechnologyvol 4 no 5 pp 325ndash330 2009

[96] M Zhou X Liang T Mochizuki and H Asanuma ldquoA light-driven DNA nanomachine for the efficient photoswitching ofRNA digestionrdquo Angewandte Chemie vol 49 no 12 pp 2167ndash2170 2010

[97] H Asanuma X Liang H Nishioka D Matsunaga M LiuandM Komiyama ldquoSynthesis of azobenzene-tethered DNA forreversible photo-regulation of DNA functions hybridizationand transcriptionrdquo Nature protocols vol 2 no 1 pp 203ndash2122007

[98] X Liang H Nishioka N Takenaka and H Asanuma ldquoA DNAnanomachine powered by light irradiationrdquoChemBioChem vol9 no 5 pp 702ndash705 2008

[99] X Liang TMochizuki andH Asanuma ldquoA supra-photoswitchinvolving sandwiched DNA base Pairs and azobenzenes forlight-driven nanostructures and nanodevicesrdquo Small vol 5 no15 pp 1761ndash1768 2009

[100] H Kang H Liu J A Phillips et al ldquoSingle-DNA moleculenanomotor regulated by photonsrdquoNano Letters vol 9 no 7 pp2690ndash2696 2009

[101] Y Zou J Chen Z Zhu et al ldquoSingle-molecule photon-fueledDNA nanoscissors for DNA cleavage based on the regulation ofsubstrate binding affinity by azobenzenerdquo Chemical Communi-cations vol 49 no 77 pp 8716ndash8718 2013

[102] Y Yang M Endo K Hidaka and H Sugiyama ldquoPhoto-controllable DNA origami nanostructures assembling into pre-designed multiorientational patternsrdquo Journal of the AmericanChemical Society vol 134 no 51 pp 20645ndash20653 2012

[103] D Han J Huang Z Zhu et al ldquoMolecular engineeringof photoresponsive three-dimensional DNA nanostructuresrdquoChemical Communications vol 47 no 16 pp 4670ndash4672 2011

[104] I A Khalil K Kogure H Akita and H Harashima ldquoUptakepathways and subsequent intracellular trafficking in nonviralgene deliveryrdquo Pharmacological Reviews vol 58 no 1 pp 32ndash45 2006

[105] S Schutze V Tchikov andW Schneider-Brachert ldquoRegulationof TNFR1 and CD95 signalling by receptor compartmentaliza-tionrdquo Nature Reviews Molecular Cell Biology vol 9 no 8 pp655ndash662 2008

[106] LW Zhang andNAMonteiro-Riviere ldquoMechanisms of quan-tum dot nanoparticle cellular uptakerdquo Toxicological Sciencesvol 110 no 1 pp 138ndash155 2009

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 22: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Journal of Nanomaterials 21

[107] A H Okholm J S Nielsen M Vinther R S Soslashrensen DSchaffert and J Kjems ldquoQuantification of cellular uptake ofDNA nanostructures by qPCRrdquoMethods vol 67 no 2 pp 193ndash197 2014

[108] J Mikkila A-P Eskelinen E H Niemela et al ldquoVirus-encapsulated DNA origami nanostructures for cellular deliv-eryrdquo Nano Letters vol 14 no 4 pp 2196ndash2200 2014

[109] M Chang C-S Yang and D-M Huang ldquoAptamer-conjugatedDNA icosahedral nanoparticles as a carrier of doxorubicin forcancer therapyrdquo ACS Nano vol 5 no 8 pp 6156ndash6163 2011

[110] M Brayman A Thathiah and D D Carson ldquoMUC1 amultifunctional cell surface component of reproductive tissueepitheliardquoReproductive Biology and Endocrinology vol 2 article4 2004

[111] S J Gendler ldquoMUC1 the renaissance moleculerdquo Journal ofMammary Gland Biology and Neoplasia vol 6 no 3 pp 339ndash353 2001

[112] C S M Ferreira M C Cheung S Missailidis S Bislandand J Gariepy ldquoPhototoxic aptamers selectively enter and killepithelial cancer cellsrdquo Nucleic Acids Research vol 37 no 3 pp866ndash876 2009

[113] S Modi C Nizak S Surana S Halder and Y Krishnan ldquoTwoDNA nanomachines map pH changes along intersecting endo-cytic pathways inside the same cellrdquoNatureNanotechnology vol8 no 6 pp 459ndash467 2013

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials

Page 23: Multifunctional DNA nanomaterials for biomedical ... · Review Article Multifunctional DNA Nanomaterials for Biomedical Applications DickYanTam 1,2 andPikKwanLo 1,2 Department of

Submit your manuscripts athttpwwwhindawicom

ScientificaHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CorrosionInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Polymer ScienceInternational Journal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CeramicsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CompositesJournal of

NanoparticlesJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

International Journal of

Biomaterials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

NanoscienceJournal of

TextilesHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Journal of

NanotechnologyHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal of

CrystallographyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

The Scientific World JournalHindawi Publishing Corporation httpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

CoatingsJournal of

Advances in

Materials Science and EngineeringHindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Smart Materials Research

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

MetallurgyJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

BioMed Research International

MaterialsJournal of

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Nano

materials

Hindawi Publishing Corporationhttpwwwhindawicom Volume 2014

Journal ofNanomaterials