SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

94
SURFACE PLASMON RESONANCE IN LIVING CELL SENSING SPR Responses in Cell-Exosome Interactions Teemu Suutari University of Helsinki Faculty of Pharmacy Division of Pharmaceutical Biosciences April 2014

Transcript of SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

Page 1: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

SURFACE PLASMON RESONANCE IN LIVING CELL SENSING − SPR Responses in Cell-Exosome Interactions

Teemu Suutari University of Helsinki Faculty of Pharmacy Division of Pharmaceutical Biosciences April 2014

Page 2: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

Tiedekunta/Osasto – Fakultet/Sektion – Faculty

Faculty of Pharmacy Laitos – Institution – Department

Pharmaceutical Biosciences Tekijä – Författare – Author

Teemu Suutari Työn nimi – Arbetets titel – Title

Surface Plasmon Resonance in Living Cell Sensing - SPR Responses in Cell-Exosome Interactions Oppiaine – Läroämne – Subject

Biopharmaceutics Työn laji – Arbetets art – Level

Master's Thesis Aika – Datum – Month and year

April 2014 Sivumäärä – Sidoantal – Number of pages

91 + 3 (Appendices) Tiivistelmä – Referat – Abstract

Surface plasmon resonance (SPR) is a label free technique to study surface interactions. It is based on photon-plasmon coupling. Laser light is directed through a prism and reflects form a metal sur-face, often gold. At certain conditions, photons turn into plasmons, which then propagate on the metal surface. The refractive index (RI) of the medium close to the metal surface alters the condi-tions when plasmons can be generated. By changing the incident angle of the light, photon-plasmon coupling can be matched. Thus, change in the SPR sensogram peak angular position (PAP) indi-cates change in the RI of the sample. Traditionally, SPR has been used to investigate biomolecule dissociation / association kinetics. Recently, it has gained popularity in living cell sensing. Exo-somes are 30-100 nm size lipid bilayer structured vesicles, which are excreted by nearly all cells. They play a role in cell-cell communications. Exosomes carry selected cargo from the cells of origin, including mRNA, miRNA, dsDNA and proteins, and they are directed to specific cells, which internalize them. This initiates responses in the recipient cells. The aim of the study was to harvest exosomes from prostate cancer (LNCaP) cells and use SPR as a novel method to detect exosome internalization by these cells. Adhesion proteins were tested in their efficiency to promote confluent cell monolayer formation on SPR gold substrate sensor sur-face. Nanoparticle tracking analysis (NTA) showed that exosome purification by ultracentrifuga-tion was successful. It was also found that gold substrate supports confluent LNCaP cell monolayer formation. Adhesion proteins did not shorten the incubation time on gold substrate, but helped the cells remain on the sensor during the SPR experiment. Prostate and platelet exosomes were tested on whether they are internalized by LNCaP cells. Control samples with plain medium and PEI/DNA nanoparticles were used. PEI/DNA particles are nonviral gene delivery vectors, which are known to permeate into cells. The SPR results showed RI increase caused 0.9 ° change in the SPR sensogram with the PEI/DNA sample and no change with the medium sample. Exosomes showed more complex responses, both increasing the PAP approximately 0.1 °. Prostate exosome sensogram returned to baseline after sample rinsing, which did not occur with platelet exosomes. It was concluded that SPR shows a response in cell-exosome interactions, which is most likely be-cause of exosome internalization. Avainsanat – Nyckelord – Keywords

Surface Plasmon Resonance, Exosomes, Prostate cancer, LNCaP cells, Cell immobilization Säilytyspaikka – Förvaringställe – Where deposited

Division of Pharmaceutical Biosciences Muita tietoja – Övriga uppgifter – Additional information

Supervisors: Lauri Paasonen and Marjo Yliperttula

Page 3: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

Tiedekunta/Osasto – Fakultet/Sektion – Faculty

Farmasian tiedekunta Laitos – Institution – Department

Farmaseuttiset biotieteet Tekijä – Författare – Author

Teemu Suutari Työn nimi – Arbetets titel – Title

Surface Plasmon Resonance in Living Cell Sensing - SPR Responses in Cell-Exosome Interactions Oppiaine – Läroämne – Subject

Biofarmasia Työn laji – Arbetets art – Level

Pro gradu Aika – Datum – Month and year

Huhtikuu 2014 Sivumäärä – Sidoantal – Number of pages

91 + 3 (liitteet) Tiivistelmä – Referat – Abstract

Pintaplasmoniresonanssi (Surface Plasmon Resonance, SPR) on leimavapaa tekniikka pintailmiöi-den tutkimiseen, joka perustuu fotoni-plasmoni pariutumiseen. Laservalo heijastuu prisman läpi metallipinnasta, joka on yleisimmin kultaa. Tietyissä olosuhteissa fotonit muuttuvat plasmoneiksi, jotka etenevät metallipinnalla. Metallin lähellä olevan väliaineen taitekerroin (Refractive Index, RI) muuttaa olosuhteita, jossa plasmoneita voi syntyä. Muuttamalla valon heijastekulmaa, voidaan aikaansaada fotoni-plasmoni pariutuminen. Näin muutos SPR sensogrammin maksimikulman si-jainnissa (Peak Angular Position, PAP) kertoo näytteen taitekertoimen muutoksesta. Perinteisesti SPR:ää on käytetty biomolekyylien sitoutumis- / vapautumiskinetiikan tutkimiseen. Viimeaikoina se on kasvattanut suosiotaan elävien solujen tutkimuksessa. Eksosomit ovat 30-100 nm kokoisia lipidikaksoiskerroksisia vesikkeleitä, joita erittävät lähestulkoon kaikki solut. Ne toimivat solujen välisessä kommunikaatiossa ja sisältävät muun muassa lähtösoluista peräisin olevaa mRNA:ta, miRNA:ta, dsDNA:ta ja proteiineja. Eksosomit kohdennetaan tietyille soluille, jotka ottavat ne sisäänsä, ja niiden soluun otto aiheuttaa vasteen näissä soluissa. Tutkimuksen tavoite oli kerätä eksosomeja eturauhassyöpäsoluista (LNCaP solut) ja käyttää SPR:ää uutena menetelmänä havaitsemaan eksosomien soluun ottoa näillä samoilla soluilla. Ko-keilimme adheesioproteiinien tehokkuutta edistää yhdenmukaisen solukerroksen muodostumista kultapinnoitteisen SPR sensorin pinnalle. Nanopartikkeli seuranta-analyysi (Nanoparticle Tracking Analysis, NTA) osoitti, että eksosomien eristys ultrasentrifugaatiolla onnistui. Totesimme myös, että LNCaP solut kykenevät kasvamaan yhdenmukaisena kerroksena kultapinnoitteisen sensorin päälle. Adheesioproteiinit eivät lyhentäneet inkubaatioaikaa kultapinnalla, mutta auttoivat soluja pysymään kiinni sensorissa SPR kokeen aikana. Testasimme LNCaP -solujen kykyä ottaa sisäänsä eturauhas- ja verihiutale-eksosomeja. Verrokkinäytteinä käytimme liuosta, joka ei sisältänyt näy-tettä ja PEI/DNA nanopartikkeleita sisältävää liuosta. PEI/DNA partikkelit ovat ei-viraalisia gee-nivektoreita, joiden tiedetään kulkeutuvan solujen sisään. Tulokset osoittivat taitekertoimen muu-toksen aiheuttavan 0,9 ° muutoksen SPR sensogrammissa PEI/DNA näytteellä. Näytettä sisältämä-tön liuos ei aiheuttanut lainkaan muutosta, kun taas eksosomit osoittivat monitahoisia muutoksia sensogrammeissa. Molemmat eksosominäytteet nostivat PAP arvoa 0,1 °. Eturauhaseksosomien sensogrammi palasi lähtöarvoon, kun näyte huuhdeltiin pois. Tätä ei tapahtunut verihiutale-eksosomeilla. Totesimme että SPR havaitsee solujen ja eksosomien vuorovaikutusten aiheuttamia muutoksia, jotka todennäköisesti johtuvat eksosomien soluun otosta. Avainsanat – Nyckelord – Keywords

Pintaplasmoniresonanssi, Eksosomit, Eturauhassyöpä, LNCaP solut, Solujen kiinnittäminen Säilytyspaikka – Förvaringställe – Where deposited

Farmaseuttisten biotieteiden osasto Muita tietoja – Övriga uppgifter – Additional information

Ohjaajat: Lauri Paasonen ja Marjo Yliperttula

Page 4: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

TABLE OF CONTENTS

ABBREVIATIONS 1 INTRODUCTION ....................................................................................................... 1 2 SURFACE PLASMON RESONANCE ...................................................................... 2

2.1 Theoretical basis of surface plasmon resonance .................................................. 2 2.2 Experimental basis of surface plasmon resonance............................................... 5

3 SURFACE PLASMON RESONANCE IN CELL SENSING .................................... 8 3.1 Instrumentation .................................................................................................... 8 3.2 Additional tools in cell sensing .......................................................................... 10

4 BIOLOGICAL APPLICATIONS WITH CELLS ON GOLD SUBSTRATE .......... 13 4.1 Cell adherence and viability .............................................................................. 13 4.2 Cell size alterations and morphological changes ............................................... 19 4.3 Intracellular activity ........................................................................................... 23 4.4 Cell-particle interactions and particle uptake .................................................... 27

5 OTHER BIOLOGICAL APPLICATIONS ............................................................... 31 5.1 Cells not immobilized to gold substrate ............................................................ 31 5.2 Bacterial cell assays ........................................................................................... 34

6 CONCIDERATIONS AND PROSPECTS OF SURFACE PLASMON RESONANCE IN CELL SENSING ......................................................................... 34

6.1 Temperature ....................................................................................................... 34 6.2 Idiosyncrasy in living cell sensing ..................................................................... 35 6.3 Fourier transform infrared and waveguide mode .............................................. 38 6.4 Surface plasmon resonance as diagnosis tool .................................................... 40

7 CONCLUSIONS ....................................................................................................... 42 8 INTRODUCTION TO EXPERIMENTAL WORK .................................................. 43 9 AIM OF THE STUDY .............................................................................................. 44 10 MATERIALS AND METHODS .............................................................................. 45

10.1 Overview of the study ........................................................................................ 45 10.2 LNCaP and PC-3 cell culture ............................................................................. 45 10.3 Exosome collection form LNCaP cells .............................................................. 46 10.4 Protein amount determination ............................................................................ 47

10.4.1 Sample washing .......................................................................................... 47 10.4.2 Protein assay ............................................................................................... 47

10.5 Exosome particle size distribution ..................................................................... 48 10.6 Gold substrate surface modifications ................................................................. 48 10.7 Immobilization of cells on gold substrate .......................................................... 49 10.8 Sample preparation for surface plasmon resonance........................................... 49

10.8.1 Exosome samples ........................................................................................ 49

Page 5: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

10.8.2 PEI/DNA polyplex samples ........................................................................ 50 10.9 Surface plasmon resonance experiments ........................................................... 50

11 RESULTS AND DISCUSSION ................................................................................ 52 11.1 Exosome yield, particle count and size distribution .......................................... 52 11.2 Cell immobilization on gold sensor ................................................................... 53

11.2.1 LNCaP cells ................................................................................................ 53 11.2.2 PC-3 cells .................................................................................................... 59

11.3 Surface plasmon resonance experiments ........................................................... 60 11.3.1 Initial scan ................................................................................................... 60 11.3.2 Peak angular position and total internal reflection area angle .................... 61 11.3.3 Peak minimum intensity ............................................................................. 66 11.3.4 Concluding remarks .................................................................................... 70

12 FUTURE PROSPECTS ............................................................................................. 74 13 CONCLUSIONS ....................................................................................................... 74 REFERENCES ............................................................................................................... 75

APPENDICES

APPENDIX 1 Medium and PEI/DNA sensograms for PC-3 cells. APPENDIX 2 Control sensograms for PAP, PMI and TIR angle. APPENDIX 3 Initial signal shifting sensograms.

Page 6: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

ABBREVIATIONS

dsDNA Double-stranded DNA

FTIR Fourier transform infrared

LNCaP Lymph node cancer of prostate cell line

mRNA Messenger RNA

miRNA Micro RNA

NTA Nanoparticle tracking analysis

PAP Peak angular position

PBS Phosphate buffered saline

PC-3 Prostate Cancer cell line

pDNA Plasmid DNA

PEI Polyethyleneimine

PMI Peak minimum intensity

RI Refractive index

siRNA Small interfering RNA

SP Surface plasmon

SPR Surface plasmon resonance

SPRI Surface plasmon resonance imaging

TIR Total internal reflection

TIRFM Total internal reflection fluorescence microscopy

Page 7: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

1

1 INTRODUCTION

Surface plasmon resonance (SPR) is a label free method for monitoring surface interac-

tions. It was first demonstrated as a way to study phenomenons occurring at metal sur-

faces in the early 1980’s (Gordon and Ernst 1980). Since then, it has gained popularity

for use in biomolecular interaction studies. Here, the other molecule is fixed on top of a

metal surface and the other molecule is introduced as an analyte. These studies include

protein-protein, lipid-protein, carbohydrate-protein, DNA-protein, DNA-DNA and

RNA-DNA interactions (Brockman et al. 1999; Nelson et al. 2001; Nelson et al. 2002;

Smith et al. 2003; Wegner et al. 2003; Mannelli et al. 2006; Yuk et al. 2006; Li et al.

2006; Wakao et al. 2008; Wong et al. 2008; Stahelin 2013). Surface plasmon resonance

has the advantage of detecting changes at the metal-biomolecule interface in real time

without labels, providing affinity and kinetic information of these processes. The tech-

nique is based on surface plasmon resonance phenomenon occurring at the metal sur-

face that is sensitive to refractive index (RI) changes at the vicinity of this surface. Mo-

lecular interactions or mass variations near the surface alter the refractive index and

thus, can be detected by SPR. Recently, this method has been applied also in living cell

sensing. Traditionally in vitro cell assays have relied on labeling methods to investigate

cell reactions to different stimulus, such as drug internalization or apoptosis (González

and Negulescu 1998; Wang et al. 2008). These methods are often time consuming and

costly. In addition, labeling may affect normal cell behavior, have limited detection

time and are usually capable of providing only end point data (Johnson 2005). As sur-

face plasmon resonance circumvents these restrictions and provides a tool to examine

cell reactions in an environment that is physiologically relevant, it has raised great in-

terest in the field of cell sensing research.

In this literature review, recent studies utilizing surface plasmon resonance technology

for living cell sensing are introduced. The focus will be on the experiments that employ

immobilization of cells on top of the sensor surface. Other assay set-ups are also dis-

cussed briefly. The aim is to shed light on the requirements and limitations that SPR has

Page 8: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

2

on cell sensing and how these limitations could be overcome. In the experimental sec-

tion, SPR is used for detection of internalization of platelet and prostate cell derived ex-

osomes by prostate cancer cells. In addition, the feasibility to culture these cells on top

of the metal surface, a prerequisite of SPR assay, is evaluated.

2 SURFACE PLASMON RESONANCE

2.1 Theoretical basis of surface plasmon resonance

Surface plasmon is an electromagnetic wave propagating at the interface between a

metal and a dielectric (Raether 1986; Maier 2007). Maxwell´s theory of surface plas-

mons describes the free electrons of a metal as an electron liquid of high density (i.e.

plasma) and density fluctuations on a surface of this liquid as surface plasmons. Plas-

mons can also be thought as the particle name of this electromagnetic wave that has

mass and velocity. Surface plasmons are not present inherently on these interfaces. In-

stead they can be created via excitation by e.g. light. Excitation of surface plasmons was

first reported by Otto (1968) and Kretschmann and Raether (1968). Kretschmann con-

figuration uses plane polarized collimated light that undergoes total internal reflection at

a metal surface.

When light is directed through a prism perpendicular to a flat surface, all light will pass

through. When the angle of incidence (θ) (the angle between the line perpendicular to

the surface and the light beam) is then increased to a critical value, the light is complete-

ly reflected and no light passes through the flat surface anymore (Figure 1). The angle

where this happens depends on the refractive index of both sides of the surface. This

angle of incidence is called total internal reflection (TIR) angle.

Page 9: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

3

Figure 1. Light travels through a prism and meets an interface where the refractive in-dex is different on both sides of the plane of interface. a) Angle of incidence (θ) is low and light passes through the plane and refracts towards the plane of interface. b) Total internal reflection (TIR). The incident angle (θ) exceeds the critical angle and all light is reflected from the plane of interface back to the prism and no light passes through the plane.

Even though at TIR no light passes through the flat surface, the electric field of the pho-

tons extends past this surface. If this flat surface is metal such as gold, photons can in-

teract with the free electrons of the gold layer. The interaction between the electric field

of the photons and free electrons of the gold depend on the momentum of the photons

and surface plasmons that are generated. For interaction (i.e. surface plasmon reso-

nance) to occur, the photon momentum must match that of the plasmons.

The momentum of photons and plasmons can be expressed as a wave vector function.

Using Maxwell´s equations these functions can be resolved mathematically into com-

ponents parallel and perpendicular to the surface. The surface plasmon wave vector

function consists of complex dielectric permittivity of the dielectric that is the medium

next to the metal, denoted by ε1 and the complex dielectric permittivity of the metal,

denoted by ε2. The vector function’s surface parallel component (kx,SP) and perpen-

dicular component (kz,SP) can be expresses as:

Equation 1. k , = ωc

ε εε + ε ,

Page 10: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

4

Equation 2. k , = ωc

εε + ε

where ω and c are the angular frequency and the speed of light in vacuum, respectively.

For photon-plasmon interaction, only the parallel vector component kx,SP of surface

plasmons matters. This is because plasmons are confined to the plane of the gold film.

For plane polarized light under TIR conditions, wave vector projection of the photons

on the metal surface kx can be expressed as:

Equation 3. k = ωc ε sin θ

where θ is the incident light angle and ε0 is the permittivity of the prism. From this

equation we can see that the plane polarized light wave vector depends on the incident

light angle θ. Wave vector coupling (i.e surface plasmon resonance) takes place when

photon wave vector projection on the metal kx equals the parallel vector component

kx,SP of surface plasmon:

Equation 4. k = k , ,

Equation 5. ωc ε sin θ = ω

cε ε

ε + ε

Page 11: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

5

2.2 Experimental basis of surface plasmon resonance

Surface plasmon-photon coupling

For surface plasmon resonance to occur, a coupling method between the surface plas-

mons and photons is required. In Kretschmann configuration, the prism itself acts as a

coupler (Figure 2). The metal surface is positioned next to the prism and surface plas-

mons are generated at the metal-dielectric interface (Homola 2006). In this type of setup

the only variable affecting the plane parallel vector component of surface plasmons

(kx,SP) is the permittivity of the dielectric (ε1) next to the gold layer. When vector cou-

pling takes place and surface plasmons are created, it can be seen as a noticeable drop in

reflected light intensity, as the photons turn into plasmons.

Figure 2. Kretchmann configuration. The prism acts as a coupler, allowing photons to interact with free electrons of metal layer. Light reflects at the metal-prism interface, but surface plasmons (SP) are generated at the metal-dielectric interface. ε1, ε2 and ε0 are the permittivities of dielectric, metal and prism respectively (Edited from Viitala et al. 2013).

As already mentioned, when surface plasmons are generated, they propagate along the

interface of metal surface and dielectric. Just as refracted light has an electromagnetic

field perpendicular to the metal surface, so do surface plasmons. This is the kz,SP com-

Page 12: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

6

ponent of surface plasmons, which extends to z-direction, i.e. into the dielectric. This

electromagnetic field of surface plasmons decays exponentially into the adjacent medi-

um in contact with the metal. This decaying field is called an evanescent field. The

depth to which this evanescent field penetrates (δz) has relation to the p-polarized light

and is about half of its wavelength (Kooyman 2008). This evanescent field is influenced

by the dielectric property (i.e. permittivity ε1) of the medium, to which it extends. Thus

the dielectric property of the medium that is within this evanescent field has an effect on

the momentum of surface plasmons. When the momentum of the surface plasmons

changes, it can be matched again by changing the incident angle of plane polarized

light.

In summary, we can conclude that in an experimental setting where a plane-polarized

light is directed through a prism at certain angle and undergoes total internal reflection

on a thin gold surface, the only variable that affects the excitation of surface plasmons is

the permittivity of the medium next to the gold surface. The reason for this is that it is

the only variable affecting the momentum of surface plasmons. The evanescent field

depth (δz) of surface plasmons depends upon the wavelength of the p-polarized light.

Changes only within this evanescent field induce changes in the surface plasmon mo-

mentum. Only when the momentum of reflected light matches that of the surface plas-

mons, can plasmons be created. When momentum matching happens, a sharp dip in the

reflected light intensity occurs. On the other hand, momentum of the reflected light can

be modified by changing the incident angle of the p-polarized light. Thus by monitoring

changes in the angle of minimum light intensity, changes within the medium next to the

gold surface can be detected. When the properties (e.g. composition) of the medium

change, it changes the permittivity i.e. refractive index (RI) of the medium.

SPR sensogram

SPR sensor instrument can either alter the incident light angle during the measurement

(angular scan), or measure the intensity changes at a fixed angle. First, a range of inci-

dent light angles is scanned from which a full SPR curve can be drawn that shows re-

flected light intensity variations in relation to angle changes (Figure 3). From this angu-

Page 13: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

7

lar scan it is readily noticeable that there is a distinct minimum in the reflected light in-

tensity at around incident angle of 68 °. This is the peak minimum angle.

Figure 3. A full SPR curve. Intensity (y-axis) is the intensity of reflected light and An-gle (x-axis) is the angle of incident light. At incident angle of ~68 °, marked by red cir-cle, a sharp drop in the intensity of reflected light is observed (Peak minimum angle).

In angular scan mode the whole angular range is scanned continuously and the position

of the peak minimum angle i.e. peak angular position (PAP) is monitored. The whole

angle range is measured e.g. every two seconds and the PAP is then expressed in rela-

tion to time in an SPR sensogram (Figure 4). A sensogram can be drawn also by using a

fixed angle scan. Here, the steepest slope at left from the peak minimum angle is chosen

and the intensity change at this angle is followed continuously. Intensity increases and

decreases at this angle translate to peak angular position decreases and increases, re-

spectively. The advantage of fixed angle is its faster response to intensity changes as

there is no need to alter the angle of the light. On the other hand, angular scan can moni-

tor the whole SPR curve continuously. Whichever mode used, SPR is capable of meas-

uring changes of the refractive index in the vicinity of the sensor slide in real time.

Page 14: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

8

Figure 4. An SPR sensogram: Minimum intensity angle is measured and plotted against time. At the beginning, a baseline is observed, where minimum intensity angle is con-stant at A. At the time indicated by the dashed arrow, a change occurs and the minimum intensity angle shifts to position B. This is due to change in refractive index of medium within evanescent field. At the end, minimum angle starts to shift back towards the baseline (Edited from Tudos and Schasfoort 2008).

From figure 4 it can be seen how the peak angular position remains constant at first.

Then, at the time indicated by the arrow, the refractive index of the medium within the

evanescent field changes and the peak minimum angle increases from A to B. Later, the

minimum angle starts to shift back towards its original position. This change in peak

angular position can be caused by, for example, molecules that adsorb on the gold sur-

face. Because the adsorbing molecules have different refractive index than the medium,

the average refractive index of the medium changes. When the molecules start to de-

tach, the space is occupied again with the medium and the refractive index changes

back, closer to the value of plain medium.

3 SURFACE PLASMON RESONANCE IN CELL SENSING

3.1 Instrumentation

A widely used experimental setup when utilizing SPR for living cell sensing deploys an

approach where cells are cultured on top of a gold surface. Although when considering

Page 15: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

9

surface plasmon excitation, silver would be a better option, gold is chemically more in-

ert and thus is the “gold standard” (Schasfoort and McWhirter 2008). The setup in-

cludes the optics (light source, photodetector and a prism), flow cell chamber, a system

for liquid perfusion and a sensor slide (Figure 5). The sensor is pressed against the

prism and flow cell chamber sits firmly on top of the sensor with a seal in between. The

flow cell chamber has an inlet and an outlet, so the medium surrounding the cells can be

changed by injection. The sensor has a very thin gold layer, approximately 50 nm, on

top of a glass. Surface plasmons are generated at the gold-dielectric interface where

cells reside. The evanescent field penetrates into the cells and refractive index changes

within cells can thus be monitored.

Figure 5. Experimental setup of living cell sensing. Cells are attached on top of a gold sensor. Sensor is placed against the prism and surface plasmons are generated on the gold surface, where cells are attached. Evanescent field is influenced by cell morpholo-gy alterations (Edited from Yashunsky et al. 2010b).

The flow cell chamber covers only a portion of the sensor slide. This has significance

when positioning the sensor slide so that the measurement area has a confluent cell

monolayer. The area of measurement is illuminated by two lasers, and is usually about 1

mm in diameter. These two lasers, which can have different wavelengths, are positioned

Page 16: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

10

at the top and at the bottom of the flow cell chamber. Consequently, the actual area of

measurement covers only a small area of the whole surface of the sensor slide. Figure 6

depicts a measurement area with a single flow cell chamber. Other cuvettes may include

two or more separate chambers. In cell sensing, the measurement area should have a

continuous cell monolayer to ensure that the signals originate from cells and not from

empty sensor. Alternatively, the use of e.g. SPR imaging allows to select a focused area

during the measurement, from which SPR signal is detected. Thus, in such a case a con-

tinuous monolayer is not required.

Figure 6. View of sensor slide from the top. Flow cell chamber is depicted as a crosscut. The gray represents the flow cell seal that separates the flow cell chamber from the rest of the sensor during the SPR measurement. The pink area is the flow cell chamber area that is filled with medium. The solid red circle indicates the area of measurement for the bottom laser and the dashed blue for the top laser.

3.2 Additional tools in cell sensing

Peak minimum intensity and total internal reflection area of the surface plasmon reso-

nance curve

When using the angular scan, the full angular spectrum is monitored throughout the ex-

periment. Thus, it is possible to gain additional information such as reflected light in-

tensity at the minimum peak of the SPR curve (peak minimum intensity, PMI) and

changes at the total internal reflection area (TIR area) (Figure 7).

Page 17: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

11

Figure 7. From a full SPR curve, information of the peak minimum intensity (PMI) and changes at the total internal reflection (TIR) area are available (Edtited from Viitala et al. 2013).

Even though these parameters are not commonly utilized, they can further aid in inter-

preting cellular events. The intensity at peak angular position (PMI) depends on losses

of the surface plasmons due to light scattering and absorption, i.e. the more light scat-

ters and absorbs, the less surface plasmons are generated and thus the reflected light in-

tensity increases (Viitala et al. 2013). Scattering is caused by all objects that have dif-

ferent refractive index than the surrounding liquid. Thus, in cellular assays the increase

in PMI is caused by e.g. corrugation of cell membrane and increasing concentration of

cell organelles within the evanescent field (Yashunsky et al. 2012b; Vala et al. 2013).

Viitala et al. (2013) simulated SPR curves for cellular monolayers and divided the cells

to a lower portion, reached by evanescent field, and an upper portion, outside of the ev-

anescent field. They saw that refractive index changes at the upper part of the cell layer,

that did not have an effect on PAP, showed consistent increase / decrease of TIR angle

as RI at this portion of cell layer increased / decreased. The intensity of TIR region re-

sponded to changes mainly within the evanescent field region. These were opposite to

PMI changes, namely, TIR intensity was increased / decreased as PMI decreased / in-

creased.

Page 18: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

12

Surface plasmon resonance imaging

Surface plasmon resonance imaging (SPRI), or 2D-SPR or surface plasmon resonance

microscopy, was first described by Rothenhäusler and Knoll in 1988 (Rothenhäusler

and Knoll 1988). It was first used for cell detection in 1999 by Giebel et al. (Giebel et

al. 1999). In contrast to a conventional SPR apparatus that measures an average RI

change in a given area, SPR imaging determines the intensity of reflected light with

high spatial accuracy using a CCD camera. It can produce an image of the sensor sur-

face from underneath (Figure 8). This makes it possible to select and detect multiple

points of interest on a single sensor slide. Resolution is limited by propagation length of

the surface plasmons and is in the range of sub-micron to micron (Wang et al. 2012b).

With this detection method RI on cellular level can be observed and RI changes be-

tween individual cells can be compared (Yanase et al. 2010; Iribe et al. 2010; Yanase et

al. 2012). The resolution is high enough to even differentiate RI change distribution

within a single cell (Wang et al. 2012b).

Figure 8. Image obtained with SPR imaging. Different cells and even different areas of single cells show different degree of reflected light intensity (Yanase et al. 2010).

Bright field and total internal reflection Fourier transform microscopy

SPR can be configured with an additional detection instrument such as bright field light

microscopy or fluorescence microscopy. With bright field microscopy it is possible to

examine e.g. cell monolayer integrity concurrently with the SPR signal (Yashunsky et

al. 2010b). Total internal reflection fluorescence microscopy (TIRFM) is used for fluo-

Page 19: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

13

rescent imaging of liquid/solid surfaces (Steyer and Almers 2001). Like in surface

plasmon resonance, an evanescent field is generated with limited penetration depth. Us-

ing this method together with SPR allows simultaneous information of fluorescence la-

beled molecules and refractive index changes (He et al. 2006). Moreover, surface plas-

mons actually enhance the intensity of fluorescence signal twofold.

4 BIOLOGICAL APPLICATIONS WITH CELLS ON GOLD SUBSTRATE

4.1 Cell adherence and viability

In order to study cellular reactions to different stimulus, cells have to attach on top of

the metal surface. Furthermore, they have to withstand a degree of shear stress, as many

experimental setups introduce a constant flow inside the flow cell chamber. Even if

measurements are conducted without flow, the injection of samples into the flow cell

chamber causes a flow of liquid that may displace loosely attached cells from the sur-

face of the sensor slide. Giebel et al. (1999) first demonstrated the use of SPR in study-

ing goldfish glial cell adhesion to aluminum-coated sensor slides. Other studies had al-

ready employed SPR in detecting whole cells (Watts et al. 1994; Fratamico et al. 1998).

In these studies, however, the cells were not cultured on top of the sensor slide. Instead,

cells flowed free on top of functionalized sensor surface, to which cells adhered.

When cells attach on surfaces, the cell membrane is not homogenously in contact with

the substrate. Cells attach by focal contacts 1 µm in width and 2-10 µm in length, where

the cell membrane is within 10-15 nm from the surface. Other areas form close contacts

where the membrane and the surface are separated by 30-50 nm or extracellular matrix

contacts that are at the distance of 100 nm or more (Figure 9) (Izzard and Lochner 1976;

Chen and Singer 1982; Burridge et al. 1988). Thus, even though SPR measures the re-

fractive index only to an extent of a few hundreds of nanometers and cell coverage on

sensor surface would be complete, a degree of extracellular fluid is within reach of the

Page 20: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

14

evanescent field. Thus, SPR always detects the effective mean of the refractive index

within the evanescent field that includes extracellular fluid.

Figure 9. Representation of cell-substrate interaction with focal-, close- and extracellu-lar matrix contacts (Edited from Giebel et al. 1999).

The degree of attachment is dependent on the cell line. Some cell types are more prone

to detach, while some do not attach at all. Also, understanding the effects that cell adhe-

sion, migration and displacement have on the SPR signals is paramount when differen-

tiating these responses from other cellular processes. Wang et al. (2012b) studied cell-

substrate adhesion strength by introducing extracellular osmotic alteration and measur-

ing the intensity changes at fixed angle. By exploiting the SPR imaging method, they

were able to obtain spatial information of reflected light intensities and thus determine

RI change distribution within single cells. They used human epithelial cell line SH-EP1

grown on top of a gold sensor that was treated with collagen to enhance cell adhesion.

Hypertonic solution induces intracellular fluid to flow out of the cell causing cell

shrinkage. This resulted in parts of the cell bottom membrane to detach from the sur-

face, seen as immediate decrease of reflected light intensity i.e. decrease of PAP. The

opposite was seen when introducing cells to hypotonic environment. Angular position

increased, as the cells were pushed towards the surface.

Yashunsky et al. (2010b) combined conventional optical microscopy to SPR instrumen-

tation to obtain simultaneous SPR information and optical images of the cell monolayer

Page 21: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

15

on the sensor slide. They monitored MDCK cells (Madin Darby canine kidney) for cell

sedimentation and attachment to the sensor surface, cell spreading, formation of cell-

cell contacts and formation of a continuous cell monolayer. The SPR measurement was

initiated by using a sensor without cells. Later, they injected the cells and monitored the

SPR response continuously. Refractive index increased rapidly as cells sedimented on

the sensor (Figure 10). Later, the RI increase is slower but reaches even higher values as

cells spread and start forming cell-cell junctions. Eventually, RI reaches its maximum

when cells occupy the whole sensor surface area and no bare gold surface is visible. In

addition to increased angular position as RI increases, the SPR curve dip became wider

and shallower. Fang et al. (2006) disturbed cell adhesion of human epidermoid carci-

noma cells (A431) by adding vincristine, a compound that inhibits microtubule assem-

bly and thus reduces cell adhesion and spreading. This disruption suppressed initial and

total increase of angular position by ~20 %. These results indicate, that by monitoring

angular position and the shape of the full SPR curve, evaluation of cell monolayer in-

tegrity can be made.

Figure 10. Refractive index change as sensor surface cell coverage increases. y-axis shows refractive index increase and x-axis is time (Edited from Yashunsky et al. 2010b).

Figure 11 illustrates single cell sedimentation on a gold sensor surface. The surface

plasmon evanescent field first senses only pure gold when cells flow free in medium.

As they make contact with the surface and consequently start to spread, SPR detects an

Page 22: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

16

increasing portion of the cell membrane and thus PAP increases as cell-substrate area

increases.

Figure 11. Schematic drawing of a cell deposition on top of a gold sensor. As cell makes contact with the sensor surface and further spreads, more of the cell surface area is within surface plasmon penetration depth (δz) (Edited from Yashunsky et al. 2010b).

Yanase et al. (2007b) correlated the amount of cells on a gold sensor to peak angular

position using rat mast cells RBL-2H3. They observed different concentrations of cells

on the sensor surface simultaneously with light microscopy and SPR. Linear correlation

was found with increasing amount of cells causing an increase in peak angular position

expressed as ∆PAP = 0.000442 × cell number mm⁄ . Ziblat et al. (2006) have also

generated an equation that enables to determine the percentage of cell coverage on a

sensor slide from the fixed angle SPR sensogram (Equation 6). They used human epi-

thelial ovarian carcinoma cells (HeLa) along with phase contrast microscopy.

Equation 6. 𝑅 = (1 − 𝑃) × 𝑅 + 𝑃 × 𝑅

where RI and RII are the reflectivities of an empty sensor and a sensor with full cell cov-

erage, respectively, R is the reflectivity and P is the degree of sensor confluency.

To further correlate PAP changes to monolayer integrity Yashunsky et al. infected

MDCK cells with Escherichia coli and used calcium-deprived medium (Yashunsky et

al. 2012a; Yashunsky et al. 2013). For E. Coli test, they utilized TIRF microscopy and

confocal imaging to observe intracellular and extracellular fluid movements, and for

calcium test, light microscopy was used. E. Coli causes cell dissociation from the sensor

Page 23: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

17

surface by modifying the focal adhesions and causes intercellular gap formation (Shifrin

et al. 2002). Calcium-deprived medium disturbs cell-cell junctions but does not affect

cell-substrate junctions (Castillo et al. 1998). In both cases, a decrease in RI was seen.

This was attributed to the compromised monolayer integrity, through which extracellu-

lar medium infiltrates the cell-substrate interface. The effect of calcium depletion is re-

versible and RI returned to the original value when the integral monolayer was re-

formed as calcium was again added.

Yashinsky et al. (2012b) extended the interpretation of the SPR response for cell cover-

age by monitoring changes not only in angular position but also in the intensity changes

at incident angle (peak minimum intensity, PMI). They monitored MDCK cell sedimen-

tation and monolayer formation, and correlated PMI variations to monolayer formation

(Figure 12). At the beginning, when no cells are present, PMI is very low. When indi-

vidual cells start to attach and spread on the surface, PMI increases proportionally to the

cell coverage. When no more cells sediment on the surface but cells occupy more sur-

face area and form cell-cell connections and small cell clusters, PMI remains constant.

Later the intensity rises rapidly. Yashunsky et al. explained this as a formation of pro-

trusions by few cells within cell clusters that generate more scattering surface. At the

final stage, the cell-medium interface is reduced as the cell monolayer is completed.

This results in PMI decrease, but still remaining higher than for the cell-free sensor.

These responses were verified by simultaneous PAP monitoring and with optical mi-

croscopy. These intensity variations were further evaluated by using human melanoma

cells (ME 1106). A confluent cell monolayer was exposed to chlorpromazine, a drug

that causes corrugation of the basal membrane of the cell i.e. membrane at the cell sur-

face interface. Disruption of the basal membrane causes more extracellular medium to

flow between the cell membrane and the substrate. This resulted in a decrease in the re-

fractive index as a result of increased water content, and also an increase in the peak

minimum intensity as a result of increased cell-medium interface at the detection area.

Page 24: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

18

Figure 12. PMI changes in relation to cell coverage. At the beginning, cells sediment on surface and cell coverage increases. Then, cells attach to each other forming cell clus-ters. Finally, a continuous monolayer is formed (Edited from Yashunsky et al. 2012b).

Cell viability on a sensor is essential for successful experimentation with cells.

Kosaihira et al. (2008) and Nishijima et al. (2010) studied the SPR response to cell

apoptosis. They used several different pancreatic cancer cell types (Hep G2, MIA PaCa-

2, PANC-1 and Suit-2) with different cancer drugs and drug combinations. They com-

bined fluorescence microscopy and SPR in one device and validated the SPR response

by monitoring the mitochondrial membrane potential, an indication of cancer drug ef-

fect on cells. The SPR response was clearly seen as a decrease in PAP as cells died.

Maltais et al. (2012) also studied SPR responses for cell apoptosis using several endo-

thelial cell types (EA.hy926, AD-293 and HeLa). Cell death was initiated with different

components that induced apoptosis by caspsase activation by extrinsic and intrinsic

pathways. This leads to cell morphology changes such as rounding, nuclear condensa-

tion and fragmentation, membrane blebbing and cell detachment. Results were con-

firmed using phase contrast microscopy and a conventional apoptosis assay with annex-

in V. Results were congruent with those by Kosaihira and Nishijima: a decrease in RI as

cells died. Maltais et al. concluded that by monitoring angular position, it was possible

to obtain accurate information of latency, rate and extent of cell apoptosis.

Page 25: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

19

Yanase et al. (2007a) demonstrated the SPR capability to measure non-adherent cells on

gold sensor by anchoring cells with different linker molecules. They employed different

coating agents on the gold sensor slides (amino-alkanethiol, dithio-

bis[succinimydylpropionate] (DSP) and a commercial agent, Sunbright). On these mod-

ified sensors they then placed human peripheral blood basophils and B-lymphocytes

that would attach to the sensor within 20 min. They observed that the SPR signal was

not dramatically different compared to adherent cells. Thus, SPR can be used for cell

sensing even if cells are non-adherent or for cells that might otherwise require extended

incubation time in order to attach and show unwanted proliferation during the culturing.

In many studies there is no need for adhesion promoters and cells adhere on bare gold

substrate strong enough. However, some researchers add adhesion proteins before cell

seeding. There is no established procedure for the use of these compounds in SPR ex-

periments and each assay must be validated separately depending on cell type and test

method used. There is no reason to believe adhesion promoters would have major effect

on the SPR signal, as these compounds are relatively small (Wang et al. 2012b). As in

some experiments these adhesion promoters are employed, however, it must be remem-

bered that these substances might adhere the investigated compounds as well, even if

the bare gold substrate would not. In many studies, the nonspecific binding is accounted

for by the use of reference sensor with bare gold and / or gold with adhesion promoter

(Kosaihira and Ona 2008; Nishijima et al. 2010; Wang et al. 2012a; Mauriz et al. 2014).

Different cells reach confluency at different rates and incubation times on the gold sub-

strate used for SPR measurements varies considerably, i.e. from 20 minutes to 7 days

(Ziblat et al. 2006; Yanase et al. 2007a). Of course depending on assay set-up, either

complete coverage or partial coverage is required.

4.2 Cell size alterations and morphological changes

The cell membrane is a semi-permeable barrier allowing fluid exchange between intra-

cellular and extracellular phases. Non-isotonic medium thus causes size alterations in

cells. MDCK and NRK-52E (normal rat kidney) cell responses in SPR were studied for

their size variations in these conditions (Robelek and Wegener 2010; Baumgarten and

Page 26: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

20

Robelek 2011; Vala et al. 2013). An increase in angular position was observed within

hypertonic medium and decrease within hypotonic environment (Figure 13). In hyper-

tonic environment cells shrink and in hypotonic they expand. Responses correlated di-

rectly to the degree of increase/decrease in medium osmolarity. The increase in PAP as

cell size is reduced in hypertonic environment conflicts that of decreasing PAP for cells

that actively shrink due to cytoskeleton contraction or inhibition of cellular respiration

(Cuerrier et al. 2008; Chabot et al. 2009). This is not contradictory though, because the

mechanisms involved for cell size shrinkage are different. When cell cytoskeleton acti-

vates, it causes morphological changes by contracting the cell. When this happens the

area occupied by the cells on gold surface decreases. Thus, SPR “sees” more of the

lower RI water content. On the contrary, when in hypertonic environment, water es-

capes from the cytoplasm of the cell but no active cytoskeleton remodeling occurs. De-

spite the reduction of cell size, RI of the cell increases due to lower water content. The

opposite holds true with hypotonic stimuli. Any bulk effect was ruled out by adding

polyvinylpyrolidone (PVP) that resulted in a bulk increase of RI on an empty sensor but

would not show response on cell covered sensor slide (Robelek and Wegener 2010).

Also, altering the osmolarity when cells were fixed did not result in any SPR response

(Baumgarten and Robelek 2011). Furthermore, the SPR data was fitted to a kinetic

model that was developed from the results obtained by optical microscopy observation

of non-isotonic cell agitation. This model correlated the cell size variations to osmotic

changes. The SPR results showed a direct correlation.

Page 27: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

21

Figure 13. PAP sensograms of MDCK and NRK cells to hypertonic (+20 and +45 mOsm/kg) and hypotonic (-23 and -39 mOsm/kg) environment. Black arrow indicates beginning of and gray arrow the end of non-isotonic stimulation (Edited from Baum-garten and Robelek 2011).

Results in hypotonic solution for MDCK and NRK cells showed more complex re-

sponses though, as initial increase in PAP before signal settled to lower values (Robelek

and Wegener 2010; Baumgarten and Robelek 2011). Similar initial opposite signal was

not visible within hypertonic medium for MDCK but did occur with NRK cells (Figure

13). This rules out the possibility of regulatory cell response, because while these cells

are capable to resist cell expansion actively by contracting, no active cell expansion is

reported (Steltenkamp et al. 2006). In addition, after reaching the minimum angular po-

sition after hypotonic stimulation, PAP showed a gradual increase while still in hypo-

tonic environment. These responses were further elucidated by inhibiting regulatory

volume changes in cells. This resulted in disappearance of gradual PAP increase but did

not affect the fast initial opposite spikes. Long-range surface plasmon (LRSP) was then

used to probe the NRK cells in both hypertonic and hypotonic solutions (Vala et al.

2013). LRSP uses enhanced optics to extend the surface plasmon evanescent penetra-

tion depth up to 1500 nm, whereas the normal penetration depth of SPR is about 200-

300 nm (Homola 2006). LRSP abolished the initial spikes completely while also the

magnitude of the response was several times stronger (Figure 14) (Vala et al. 2013).

This reveals a very essential property of surface plasmons in cell sensing. Deeper probe

Page 28: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

22

depth measures the same cell-surface interface as regular surface plasmon with few

hundred nm penetration depth, but also much of the cell cytoplasm. Thus, LRSP signal

is less influenced by changes occurring at cell-surface interface. For regular surface

plasmon, the pronounced spikes were thus explained as events occurring at the basal

cell membrane.

Figure 14. Long-range surface plasmon resonance sensogram of NRK cells under hypo-tonic stimulation. Initial opposite spikes are completely abolished (Edited from Vala et al. 2013).

Wang et al. (2012b) used human epithelial cells (SH-EP1) to conduct similar experi-

ments by altering the osmolarity of medium. They employed an SPR imaging method

with the usual penetration depth of 200-300 nm, but with the capability to determine

PAP shift at very high spatial accuracy. Thus, they could measure distinct areas of sin-

gle cell instead of the “effective mean” of refractive index within a cell monolayer.

They also observed initial opposite spikes with both hypertonic and hypotonic stimula-

tion. Some areas of the cell showed smaller initial spike effect, while other areas larger.

These localized responses were attributed to vertical movement of cell membrane as

osmotic pressure either detached parts of the cell membrane from the surface or pushed

them toward it.

Page 29: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

23

These results come to show how important it is to know the properties of used instru-

mentation and the mechanisms that different stimulus have on the cells; similar cellular

response may lead to opposite signal in SPR or similar response in SPR can be attribut-

ed to different mechanism within the cells.

4.3 Intracellular activity

In addition to simple cell membrane occupancy on sensor surface or intracellular fluid

amount, changes in cell membrane composition are reflected on the SPR signal as

demonstrated by Ziblat et al. (2006). They measured the cholesterol amount within the

cell membrane using cyclodextrin bound cholesterol to deliver cholesterol into the cell

membrane and cyclodextrin alone to deplete the membrane of it. Since PAP increased

with increasing cholesterol concentration, SPR was capable of detecting the degree of

cholesterol enrichment within the cell membrane. They confirmed the results by build-

ing an artificial lipid bilayer that showed the same response. Also, thin section micros-

copy confirmed that no morphological changes occurred that would have affected the

signal. Thus, the SPR was measuring the cell membrane composition alterations and not

e.g. changes in membrane thickness. Also, already in 2002 Hide et al. studied mast cell

(RBL-2H3) response to antigens and observed that SPR showed a prolonged response

when antigen was bound on cell receptors (Hide et al. 2002). They reasoned that due to

the long duration of the response, it had to originate not from a simple ligand-receptor

binding, but from some other cellular responses initiated by these substituents. Further-

more, the concentration of membrane receptor ligand did not alter the degree of SPR

signal response (Hiragun et al. 2012; Mauriz et al. 2014).

Actin cytoskeleton rearrangement is one of these cellular responses that are widely ac-

claimed to be responsible of the SPR signal. The activation of angiotensin-1 (AT1) re-

ceptor results in G-protein signal response (Cuerrier et al. 2009). Fluorescence labeling

of cell actin cytoskeleton allows monitoring of cytoskeleton rearrangement along with

refractive index changes. TIRFM-SPR shows that G-protein activation in HEK 293

(human epithelial kidney) cells leads to cytoskeleton rearrangement within the basal

membrane, resulting in increased actin density in the area (Chabot et al. 2013). Upon

Page 30: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

24

injection of AT1 receptor ligand, a rapid decrease in SPR signal is observed followed by

a gradual increase above the original value (Figure 15). This initial decrease is most

likely due to the strong negative signal caused by cell contraction and intercellular gap

formation overlaying the signal from cytoskeleton rearrangement. The prolonged in-

crease in signal is likely the result of increasing contribution of cytoskeleton rearrange-

ment to the SPR response, as cells eventually spread and close intercellular gaps, while

increased basal actin density is still present. More evidence of actin cytoskeleton contri-

bution was shown on MDCK and A7r5 (rat vascular smooth muscle) cell treatment with

cytochalasin D and Latrunculin A, respectively (Chabot et al. 2013; Michaelis et al.

2013). These compounds lead to actin cytoskeleton disassembly by inhibiting actin

polymerization, the effect of cytochalasin D, or by inducing depolymerization, the ef-

fect of Latrunculin A. Cell monolayer remains intact and cells do not detach. This cyto-

skeleton degradation resulted in permanent decrease in SPR signal in both cases.

Figure 15. RI change for human epithelial kidney (HEK 293) cells as AT1 –receptor is activated with angiotensin II (black line). Gray line is control (Edited from Chabot et al. 2013).

In addition to cytoskeleton rearrangement, other intracellular events could contribute to

SPR signal as well. Yanase et al. (2007a; 2007b) and Horii et al. (2011) stimulated

mouse keratinocytes (PAM212), basophils and RBL-2H3 mast cells with epidermal

growth factor (EGF) or different antigens. They observed cell spreading with confocal

Page 31: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

25

microscopy and differential interference contrast microscopy to relate cell spreading

and PAP changes. They showed that simple cell spreading, ruffling or cell membrane

density changes did not explain PAP increase completely and the PAP increase was still

observable even after they eliminated cell spreading by destroying the actin cytoskele-

ton. They associated PAP alterations directly to antigen or EGF binding. Furthermore,

for the cells stimulated with antigens that results in exocytosis and histamine release,

both increase and decrease of PAP response were observed. They concluded that despite

the direction of SPR response, PAP changes measure some intracellular events induced

by these stimulants. They did not, however, investigate the cause of these opposite re-

sponses. However, these stimulants induce various intracellular events such as changes

in pH, temperature and membrane potential, cytosolic protein movement to plasma

membranes or other organelles, phosphorylation / dephosphorylation or release of

membrane proteins that lead to activation of protein kinases and release of intracellular

calcium (Siraganian 2003).

Odorant molecules interact with olfactory receptors, and through G-protein activation

initiate signaling cascade, where secondary messengers such as cAMP (cyclic adeno-

sine monophosphate) and IP3 (inositol trisphosphate) are produced by action of adenyl-

ate cyclase and phospholipase C (PLC), respectively (Figure 16). These signaling path-

ways also result in an increase of intracellular calcium and the SPR signal (Lee et al.

2006). Detecting cell calcium concentration is, in fact, used as a method to measure in-

tracellular activity, and it was thought to be the direct cause also for the increased SPR

signal (Ivic et al. 2002; Auger-Messier et al. 2004; Lee et al. 2006). However, the pres-

ence of calcium was later shown not to be responsible of the SPR response (Cuerrier et

al. 2008).

Page 32: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

26

Figure 16. Intracellular signaling pathway initiated by odorant molecule binding to ol-factory receptor. Receptor activation causes phospholipase C (PLC) to initiate signal transduction, where inositol bisphosphate (PIP2) is transformed into inositol triphos-phate (IP3). IP3 causes endoplasmic reticulum (ER) to release calcium into cytoplasm. SPR detects these responses (Edited from Lee et al. 2006).

Some work has been done in an attempt to reveal the cellular signaling pathways that

are responsible for the SPR signals. VEGF (vascular endothelial growth factor) and

EGF are growth factors that induce several intracellular cascades to activate, including

extracellular regulated kinase (ERK) and phosphatidylinositol 3´-kinase (PI3K) mediat-

ed pathways (Hoeben et al. 2004; Ono and Kuwano 2006). Binding of VEGF to mem-

brane receptor results in an increase of SPR signal that is abolished by the use of con-

current antagonist (Mauriz et al. 2014). EGF stimulation results in a complex response

in SPR and only a minimal response when the EGF receptor is mutated on the ATP

binding site (K721M) (Hiragun et al. 2012). The mutation prevents the phosphorylation

in the tyrosine residues of the receptor even if EGF binds to the receptor and thus sig-

naling is inhibited. It was also shown that inhibition of PI3K protein kinase alone alters

the normal SPR response. Interpretation of this change is difficult though, as responses

to EGF show complex sensograms with increasing and decreasing PAP. Tanaka et al.

(2008) and Yanase et al. (2010) investigated intracellular signaling pathways using

RBL-2H3 mast cells and found that the activation of early intracellular signaling mole-

Page 33: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

27

cules spleen tyrosine kinase (Syk), linker for activation of T cells (Lat) and Grb2 –

related adaptor protein (Gads) and their subsequent activation of protein kinase C iso-

type β (PKCβ) are necessary for RI change. It is shown that upon activation, PKCβ is

translocated to cell membrane and thus, could be responsible of the RI change (Abdel-

Raheem et al. 2005). Yet another explanation for SPR signal was given by Kosaihira

and Ona (2008) as they measured mitochondrial membrane potential changes caused by

different anticancer drugs. They associated RI decrease directly to a decrease in this

membrane potential.

From these results, it is evident that in cells a variety of responses occur, that can be re-

sponsible for the SPR signal, and determining the actual source of the signal is difficult

and still not fully understood. The situation is even further complicated by the fact that

many different mechanisms can contribute to the SPR signal simultaneously, such as

cell spreading / contraction and several concurrent intracellular events.

4.4 Cell-particle interactions and particle uptake

Cell-particle interactions can be detected using SPR. Wang et al. (2012a) studied lectin

binding to membrane proteins of human epithelial (SH-EP1) cells and were able to cor-

relate RI increase / decrease to lectin-membrane protein attachment / detachment. It was

even possible to calculate ligand association / dissociation constants from the SPR data.

This bears significance, since membrane proteins are very often targets for drugs (Hop-

kins and Groom 2002). When using living cells for these kinetic data calculations, there

is no need to isolate membrane proteins and drug-ligand interactions can be detected in

their natural environment. This also might be valuable information in novel drug dis-

covery. Aside from simple association / dissociation, evidence of particle uptake was

shown in form of decreased dissociation constant in living cells compared to dissocia-

tion constant in dead cells. This was interpreted as an indication of cellular uptake of the

ligand.

While it might be expected that cell-particle interactions would always result in similar

responses in SPR, this is not the case. This is well described by the situation where a

Page 34: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

28

decrease in refractive index was observed, when investigating transferrin uptake with

human melanoma cells (MEL 1106), while the uptake of PEI/DNA polyplexes in HeLa

cells resulted in an increase of RI (Yashunsky et al. 2009;; Silén 2013). Transferrin is an

iron carrier that is internalized by cells by endocytosis. It can be used in enhancing the

uptake of therapeutic drugs, including large protein drugs (Qian et al. 2002). PEI/DNA

is a nanoparticle used as a non-viral gene delivery vector, and likewise uses endocytic

pathways to reach the intracellular space (Boussif et al. 1995). Transferrin was also test-

ed on an empty sensor, which resulted in only minor increase in the RI (Yashunsky et

al. 2009). Even though no mention was given on cell monolayer integrity during exper-

iments, SPR response correlated to transferrin concentration used, and inhibition of en-

docytosis abolished the signal. Thus, the decrease in RI when cells are present is unex-

pected, as if SPR is presumed to reflect merely increased concentration of internalized

particles, the refractive index should increase. It was thus concluded that the ability of

SPR to detect drug internalization does not originate from direct observation of mole-

cule presence within cell, but rather a result of increased water content of the cells as

endocytic vesicles carried extracellular fluid within. This theory is further supported by

confocal imaging and SPR experiments with Escherichia coli infected MDCK II cells

(Yashunsky et al. 2013). Upon infection, formation of endocytic vesicles transferring

extracellular medium inside the cells is observed at the basal membrane, accompanied

by reduction in RI. Even though this does not explain the opposite result for PEI/DNA,

cells internalize transferrin and PEI/DNA by different endocytic process. Transferrin

undergoes clathrin-mediated endocytosis, while PEI/DNA is taken up by fluid-phase

endocytosis (Rémy-Kristensen et al. 2001; Ehrlich et al. 2004). Although both these

processes carry extracellular fluid into cells, there are certain differences e.g. clathrin

vesicle size is ~100 nm, whereas fluid-phase vesicle size is several hundreds to thou-

sands of nm (Samaj et al. 2004). Also, transferrin is a very small molecule of about 90

kD, whereas PEI/DNA particles are several thousands of kD in size. Thus, the amount

of mass introduced into cell is massive with the PEI/DNA particles. It should also be

recognized that different cell types show different endocytosis kinetics and even differ-

ent responses to the same stimulus (Hiragun et al. 2012). In these studies, PEI/DNA up-

take was tested using HeLa cells, whereas transferrin was tested using MEL 1106 cells.

Page 35: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

29

Thus, these ambiguous results can be caused by e.g. endocytosis at different locations in

cell membrane.

Differences in SPR signal are also observed between two passively absorbing drugs,

propranolol and D-mannitol (Viitala et al. 2013). When confluent monolayer of MDCK

II cells is stimulated with these compounds, an initial decrease in RI is observed for

both. For D-mannitol, after sample injection is stopped, PAP returns to its original val-

ue. For propranolol, angular position increases past the original value (Figure 17). Fur-

thermore, this increase in PAP is concentration dependent.

Figure 17. PAP changes when MDCK II monolayer is introduced to different concen-trations of propranolol and D-mannitol. The downward arrows show injection of sample and upward arrows the end of sample injection (Viitala et al. 2013).

Even though both compounds are absorbed through cellular barriers passively, D-

mannitol uses the paracellular and propranolol the transcellular route. It was concluded

that because D-mannitol uses paracellular absorption route, it causes disruption in the

integrity of the cell monolayer as it facilitates its movement past the monolayer travel-

ing between the cells. Thus, decrease in RI results from increasing amount of water at

cell-substrate interface. Propranolol on the other hand passes into the cells. Accumula-

tion of the drug and subsequent cellular responses are seen as increase in the refractive

index.

Page 36: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

30

To further shed light in these different responses, Viitala et al. (2013) observed also the

peak minimum intensity (PMI) and the TIR area. It was revealed that for D-mannitol

the intensity remained the same regardless of concentration. For propranolol, on the

other hand, PMI followed PAP changes and was also concentration dependent. This

could indicate that propranolol initiates a mass redistribution within the cell, first away

from the evanescent field and later toward it. Thus, the eventual positive shift in angular

position and intensity increase is because of accumulation of propranolol, accompanied

by mass redistribution to the evanescent field and possibly cell spreading. Here, the

mass redistribution results from cell organelles and other constituents moving into the

evanescent field, introducing roughness and interfaces that scatter and absorb more

light. This same effect was observed by Vala et al. (2013) as cell organelle concentra-

tion increased when the fluid volume of the cells decreased. TIR area changes further

confirmed the PAP and PMI sensograms. The TIR angular position for both, proprano-

lol and D-mannitol followed the same pattern as the PAP sensograms (Viitala et al.

2013). Furthermore, the reflection intensity at the TIR area was opposite to the PMI

change for propranolol. These responses were compared to simulated cell monolayer

sensograms. The monolayer was theoretically divided into two sections: lower part

within the reach of the evanescent field (EF) and upper part outside the evanescent field

(Cell) (Figure 18). It was found that the TIR angular position increase was a result of

mass accumulation only at the upper part of cell monolayer, whereas the TIR area inten-

sity decreased as the amount of light scattering surfaces increased only at the lower part

of the cells.

Figure 18. Schematic depiction of a cell monolayer divided theoretically into lower re-gion within the evanescent field (EF) and upper region outside the evanescent field (Cell) (Edited from Viitala et al. 2013).

Page 37: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

31

From these results, we can conclude the intracellular events that are responsible for dif-

ferent SPR signals (Table 1). RI increase, or mass accumulation, at the lower part of the

cell (EF) increases PAP, whereas the same change outside evanescent field (Cell) in-

creases TIR angle (θ). The increase in the amount of light scattering surfaces within ev-

anescent field increases PMI and decreases TIR intensity, whereas when this happens

outside evanescent field, it has no effect on PMI. These parameters are not yet widely

used in cell sensing assays, and future studies including evaluation of all these SPR pa-

rameters could deepen the understanding of cellular events responsible of the SPR re-

sponses.

Table 1. Different changes at different parts of the cell monolayer cause responses in different SPR parameters. Arrows up or down indicates increase or decrease in the pa-rameter, respectively. Minus indicates no change in parameter can occur. Cell region is upper part of cell monolayer outside the evanescent field and EF region is the lower part, within the evanescent field. Data based on Viitala et al. (2013).

RI Light scattering PAP PMI TIR θ TIR in-

tensity

Cell -region

Ï −

Ï − Ï

EF -region

Ï − Ï Ð

Ï Ï − − −

5 OTHER BIOLOGICAL APPLICATIONS

5.1 Cells not immobilized to gold substrate

SPR has also found its use in assays where cells are not immobilized on a sensor. A va-

riety of detection formats have been applied that functionalizes gold substrate with mol-

Page 38: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

32

ecules that then capture cells flowing on top of the sensor slide. Methods using mono-

clonal antibodies to recognize red blood cells, lymphocyte cancer cells and T- or B-cells

have been developed (Quinn et al. 2000; Suraniti et al. 2007; Zhang et al. 2014). The

SPR response for cells attaching on sensor is cell concentration dependent. Also, even if

different cell types show similar adherence to the same ligand, different cell types show

different SPR signal. Optimizing is required though, as with increasing the cell concen-

tration non-specific binding starts to obscure the cell-ligand signal (Mauriz et al. 2014).

Bombera et al. (2012) constructed a DNA-array to capture B- and T-cells from a com-

plex cell mixture (Figure 19). The array consisted of an antibody attached to DNA

grafted on the gold sensor. Antibody captures cells specific to it and within the DNA

bridge, an intermediate DNA strand was included that could be activated enzymatically

to release the attached cells. Thus, different SPR systems for recognizing and sorting of

specific cells could potentially be fabricated.

Figure 19. Schematic drawing of a DNA-array for the capture and release of cells. a) DNA grafted on the sensor surface, b) intermediate DNA that can be activated enzymat-ically, c) antibody for recognition of the targeted cell types, d) targeted cell (Edited from Bombera et al. 2012).

Page 39: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

33

Mauriz et al. (2014) used a VEGF functionalized sensor to which cells attached non-

covalently. The sensor was regenerated by removal of attached cells with reagents. The

same functionalized sensor could be used for up to 68 times without major loss in SPR

signal. These types of assays show promise also e.g. in drug discovery. A candidate

drug targeted for membrane receptor could be tested for its ability in preventing cell ad-

hesion to a functionalized sensor surface (Mizuguchi et al. 2012). This circumvents the

need to isolate the membrane protein, which can often be challenging.

Cell secretion is also studied by using the SPR technique. Milgram et al. (2011) inte-

grated the sensor slide on the bottom of a cell culture well. B-lymphocytes placed with-

in the culture well secreted specific antibodies that then diffuse and interact with the

sensor surface functionalized with antigen proteins. A similar system was used to study

VEGF secretion of ovarian cancer cells (Liu et al. 2012). The sensor slide was function-

alized with antibody that binds to VEGF. Cells were, however, grown on top of the flow

cell chamber (Figure 20). The ability of these functionalized sensors to determine the

concentration of the ligand of interest must first be validated. After validation, determi-

nation of the amount of secreted molecules per amount of cells can be calculated with

low detection limit and fast response.

Figure 20. Schematic depiction of detection scheme with cells attached on top of the flow cell chamber. Cells secrete VEGF that is then bound to the antibody functionalized on the sensor surface. SPR detects the amount of VEGF bound to the antibody (Liu et al. 2012).

Page 40: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

34

5.2 Bacterial cell assays

In addition to mammalian cells, bacterial cells have been employed in SPR applications.

Detection of specific bacteria using surface plasmon was first demonstrated by Watts et

al. (1994), using an antigen-functionalized sensor for binding of Staphylococcus aureus

injected as analyte. Since then, many researchers working with bacteria have utilized

SPR. In these studies, Escherichia coli and S. Aureus are commonly used strains. Tawil

et al. (2012) have developed a sensor for the detection of E. Coli and methicillin-

resistant S. aureus. The sensor surface is functionalized with highly specific bacterio-

phages, enabling the device to differentiate between methicillin-sensitive and methicil-

lin-resistant strands. Another group has developed antibody-functionalized sensors that

are capable of detecting E. Coli and Salmonella spp. from milk, juice and ground meat

samples with minimal sample pretreatment (Waswa et al. 2006; Waswa et al. 2007).

The device is portable, cheap and due to the benefits afforded by surface plasmon, la-

bel-free and fast. Other researchers have used bacterial cells somewhat differently. By

fixing E. Coli on top of the sensor surface, it is possible to detect C-reactive protein

(CRP) or pollutants as they bind to or damage the bacteria, respectively (Choi et al.

2005; Lee et al. 2012). Bacteria itself can thus act as part of the detector. These kinds of

SPR devices could be used for detecting and differentiating dangerous pathogens more

quickly, as diagnostic tools for detection of inflammatory constituents from patients or

tools to detect drugs or dangerous toxins.

6 CONCIDERATIONS AND PROSPECTS OF SURFACE PLASMON RESO-

NANCE IN CELL SENSING

6.1 Temperature

The refractive index of liquids is temperature dependent. A one-degree Celsius change

causes the angular position to shift ~0.014 ° (Schasfoort and McWhirter 2008). There-

fore, temperature variations during assay must be prevented. Also, at higher tempera-

Page 41: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

35

tures photon-electron scattering increases, resulting the peak angular position to shift to

higher values, and SPR curve to become wider and shallower (Akimoto et al. 2000;

Chiang et al. 2001). All of these factors result in lower measurement accuracy.

Temperature effect on SPR responses in living cell assays has not been explicitly inves-

tigated. Nevertheless, Yanase et al. (2007a) reported in their experiment a complete dis-

appearance of PAP response when basophils were stimulated with anti-IgE at 4 °C

compared when stimulated at 37 °C. In another study done by Yashinsky et al. (2009)

similar disappearance of SPR signal was observed with human melanoma cells as they

observed transferrin-induced endocytosis at temperatures of 37 °C and 19 °C. On the

other hand when transfecting HeLa cells with PEI/DNA particles, PAP shows pro-

nounced response within lower temperatures (Silén 2013). These opposite responses

may be partially explained by their different endocytic processes. PEI/DNA is trans-

ferred into cells by non-specific fluid-phase endocytosis, while transferrin-induced en-

docytosis is clathrin-mediated process (Rémy-Kristensen et al. 2001). The clathrin in-

duced endocytosis is temperature sensitive and ceases completely at temperatures below

37 °C, whereas fluid-phase endocytosis continues even at very low temperatures

(Illinger et al. 1991). The explanation for stronger signal for PEI/DNA nanoparticles at

lower temperatures could be in the altered cellular processes and higher sensitivity of

SPR at lower temperatures.

6.2 Idiosyncrasy in living cell sensing

When comparing SPR responses in cell assays, it should be remembered that cell type

might play a pivotal role in measured response. Some cells, such as MDCK form co-

lumnar type monolayers where the apical cell membrane i.e. membrane not in contact

with the sensor surface may not be within the reach of evanescent field (Reinsch and

Karsenti 1994). Other cells, such as human epithelial cells SH-EP1 exhibit a shape

where the apical membrane distance from the surface varies (Wang et al. 2012a). In the

experiment done by Wang et al. on detecting lectin binding to the cell surface, they

were able to see an SPR response. They used SPR imaging to resolve the distribution of

RI changes within single cells and fluorescence imaging to correlate lectin binding to RI

Page 42: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

36

changes. It was evident that RI increased at the outer regions of the cell while no change

was observed at the middle. This shows that as peripheral areas of the cell exhibit flatter

morphology than the middle, the evanescent field penetrates through the whole cell

body in these peripheral areas (Figure 21). Intracellular effects were ruled out by repeat-

ing the experiments by fixing the cells. Were they to repeat the same experiment with

e.g. MDCK cells, it might have resulted in complete lack of signal, due to the shape of

these cells.

Figure 21. SH-EP1 cell shape allows the detection of ligand binding to the membrane receptor on the apical membrane only at the edge of the cell because of the limited pen-etration depth of surface plasmon (Wang et al. 2012a).

The variation between different cell types in their response to same stimulus is well

demonstrated by Hiragun et al. (2012). They used EGF, a protein that initiates a cascade

of intracellular events, to compare different cell reactions. With six different cell lines

sensitive to EGF, they observed very different signal patterns and intensities. Some cells

showed triphasic sensograms, whereas other exhibited di- or monophasic responses.

Propagation length of surface plasmons on the gold surface is in a range of 8-10 µm,

when the common 600-800 nm wavelength is used (Johansen et al. 2000). The surface

area occupied by cells varies depending on the cell size and shape. The degree of cell

occupancy on the sensor effects on whether a surface plasmon propagating on gold sur-

face detects the refractive index of a single cell, cell and medium combined or medium

only (Figure 22). When detecting cell reactions, more cell surface on detection area re-

sults in stronger SPR signals. Also, if a variation exists between experiments, result

comparability is compromised.

Page 43: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

37

Figure 22. Propagation length of surface plasmon (Lx) and degree of cell occupancy de-termines whether surface plasmon propagating on a sensor surface detects only cell sur-face a), cell surface and medium combined b) or only medium (Edited from Golosovsky et al. 2009).

Also, no attention to cell cycle has been given in any studies. Although, perhaps not so

relevant when probing a single cell, as most commonly used SPR assays detect a large

group of cells, the effect of cell cycle might be worth considering. While cells are at dif-

ferent phases in their cell cycle, differences in cellular events, such as DNA and protein

synthesis and accumulation, occur (Douglas and Haddad 2003). In different cell cycle

phase, responses on the same stimulus may differ. For example, the efficiency of many

anticancer drugs within a single cell depends on the cell cycle phase (Schwartz and

Shah 2005). Synchronizing the cell cycle might provide more similar responses between

cells and thus, more repeatable signals in SPR measurements.

There are other factors that may also play a role in the repeatability and comparability

of assays, but are rarely given much attention. The flow in SPR measurements causes a

shear stress, which can induce intracellular events that activate signaling pathways

(Chen et al. 1999; Chachisvilis et al. 2006). In addition, the passage number of cells has

an effect on cell morphology, adhesion, response to stimuli, growth rate and protein ex-

pression, all of which can have profound effect on the SPR response (Esquenet et al.

1997; Briske-Anderson et al. 1997; Lin et al. 2003). Also, incubation time on the gold

substrate and the use of adhesion proteins varies in different studies, depending on type

of assay and cells used, and are rarely considered extensively.

Page 44: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

38

6.3 Fourier transform infrared and waveguide mode

In an effort to extend the probing depth of surface plasmon technology to better suit the

needs of living cell sensing, the use of longer wavelength seems logical as penetration

depth of evanescent field correlates to it. Traditionally SPR operates within visible light

wavelength. This was extended to near- and mid-infrared range using a Fourier trans-

form infrared surface plasmon resonance device (FTIR-SPR) (Ziblat et al. 2006; Lirts-

man et al. 2008). In addition of scanning a range of incident angles, a continuum of

wavelengths is scanned simultaneously. Thus, tuning of these parameters allow maxi-

mum sensitivity and appropriate penetration depth. In addition to penetration depth, the

propagation length of SP increases along with increasing wavelength. The penetration

depth and propagation length are affected by several factors e.g. water absorption at cer-

tain wavelengths (Golosovsky et al. 2009). These parameters can be resolved mathe-

matically (Figure 23). By selecting appropriate conditions for the used cell assay, a

large portion of cell volume can be detected.

Figure 23. Penetration depth (δz) a) and propagation length (Lx) b) of surface plasmons at infrared wavelengths. Average height, membrane thickness and lateral size of an epi-thelial cell are marked (Edited from Golosovsky et al. 2009).

Golosovsky et al. (2009) theorized that with FTIR-SPR it might be possible to capture

even more information of the cell monolayer by detecting the formation of guided mode

in the SPR sensogram when using these longer wavelengths. Indeed, Yashunsky et al.

Page 45: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

39

(2010b) observed an additional reflection minimum, separate to that caused by surface

plasmon coupling (Figure 24).

Figure 24. Second reflection minimum separate from SP minimum is observed. This minimum is due to generation of guided mode that can exist when certain conditions fulfill i.e. correct cell height, monolayer integrity, correct angle of incidence and correct wavelength (Edited from Yashunsky et al. 2010b).

This additional minimum can only exist when the cell monolayer is able to support the

formation of a waveguide mode. At a certain angle and wavelength, when light meets

the interface of the substrate and the cell layer, part of the light reflects back and part

refracts into the cell layer (Figure 25) (Yashunsky et al. 2012a). When the cell height

and wavelength match, all refracted light is reflected back at the cell-medium interface

at the apical cell membrane. Further reflection and refraction takes place again at the

cell-substrate interface and thus, light travels within the cell monolayer. In order for this

waveguide to exist, cells must form an uninterrupted, tightly connected layer. When this

layer starts to break, intensity of this guided mode decreases.

Page 46: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

40

Figure 25. Schematic representation of tight cell monolayer supporting a waveguide mode. At certain angle (Θinc) and wavelength light passes to monolayer and reflects at cell-medium interface. When cells are tightly connected, light travels within the cell monolayer (Edited from Yashunsky et al. 2012a).

As can be seen from figure 24, FTIR-SPR detects reflected light intensity with varying

wavelengths. This allows the search of both the guided mode and surface plasmon

peaks, as these do not occur at the same wavelength. Transition of SP peak minimum

intensity to lower and higher wavelength corresponds to refractive index decrease and

increase, respectively. The position of the guided mode peak reflects the cell monolayer

height and intensity corresponds to cell monolayer integrity.

6.4 Surface plasmon resonance as diagnosis tool

As already mentioned throughout this review, the use of SPR in living cell sensing may

open new possibilities for tools in diagnostics and drug development. Allergy screening

can be performed by culturing basophils, obtained from patients, on a sensor surface

and introducing different allergens to them. Cell response in the SPR reveals to which

allergens cells react (Suzuki et al. 2008). In some cases, cells may not release histamine,

even if an allergen causes a reaction in them. Thus, conventional allergy tests would not

identify these patients, whereas SPR does. Figure 26 shows the SPR response of baso-

phils obtained from a pollen allergy patient to different allergens (Yanase et al. 2012).

IgE -antibody, mite- and cedar pollen antigens provoke a clear response, whereas sweat

antigen does not. For a patient with atopic dermatitis sweat antigen does show a re-

Page 47: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

41

sponse. Using SPR imaging, a multi-well array could be constructed for high through-

put screening, as SPRI is capable of monitoring refractive index changes in each well

simultaneously (Yanase et al. 2013).

Figure 26. Sensograms of response to different allergens by basophils obtained from allergy patient with pollen allergy (Edited from Yanase et al. 2012).

With SPR, sorting of different cells from a heterogeneous mixture is possible. Biosensor

functionalized with antigens specific to different lymphocytes separates B- and T- cells

(Suraniti et al. 2007). On the other hand, when a mixture of cells is cultured on a gold

surface, SPR imaging can differentiate individual cells that react when antibody specific

to them is added (Iribe et al. 2010). Even cells at different stages of differentiation can

be identified according to their distinct responses (Mir and Shinohara 2012). Also,

screening of cancerous cells from a mixture of several cells is possible, as shown by

Hiragun et al. (2012). Prostate cancer cells showed a unique response to EGF. Thus,

with SPR imaging method even a single cancerous cell could be identified from a large

population.

In drug development, SPR could speed up the analysis of new candidate drugs. It is

possible to evaluate therapeutic potential of new cancer drugs and drug combinations in

their time-course and degree of apoptosis on cells (Kosaihira and Ona 2008; Nishijima

et al. 2010). Also, a new method for evaluation of the absorption mechanism of drugs

Page 48: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

42

through cellular barriers could be possible, as different absorption mechanisms can be

separated by their characteristic effects on peak angular position as well on peak mini-

mum intensity (Viitala et al. 2013). With SPR, these assays could be performed within

minutes as opposed to hours or days required with the currently used methods.

7 CONCLUSIONS

In living cell assays, the degree of cell coverage and cell viability on sensor surface is

essential and must be addressed for each assay. As SPR signal is sensitive to the amount

of water within the sensing area, varying cell occupancy on sensor results in differences

in SPR signals. Consequently, results become unreliable and incomparable. A simple

method to ensure cell coverage is to observe the sensor slide before SPR experiment

using microscopy. Also, the integrity of the cell monolayer can be evaluated from a full

SPR curve. Some SPR instruments allow monitoring of PMI in addition to PAP, which

can further aid in cell monolayer integrity assessment. Furthermore, additional tools

such as SPR imaging can be used.

Cell reactions can be seen as responses in SPR sensograms. However, the cause of the

response can be difficult to interpret. PAP detects RI changes within the evanescent

field, but many different cell reactions can cause these changes. Moreover, when these

reactions happen simultaneously, RI changes can overlay each other. In this case, sig-

nals can become impossible to interpret and to link to specific cell responses. For this

reason, cell assays must be designed so that SPR response can be attributed to a specific

cell reaction of interest. This can include the use of control experiments or appropriate

additional instrumentation, such as confocal microscopy, TIRFM or FTIR-SPR. In addi-

tion, other SPR parameters such as PMI and TIR area changes can help in understand-

ing the origin of these responses.

Page 49: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

43

8 INTRODUCTION TO EXPERIMENTAL WORK

Exosomes are lipid bilayer structured vesicles of 30-100 nm in diameter. Other extracel-

lular vesicles, such as microvesicles and shed vesicles can overlap in their size, but usu-

ally they are larger than exosomes (Cocucci et al. 2009; Akers et al. 2013). However,

exosomes from prostate origin, often called prostasomes, have been reported to be up to

150 nm in diameter (Llorente et al. 2007; Jansen et al. 2009; Witwer et al. 2013). No

definitive criteria yet exist to differentiate exosomes from these other vesicles. Differen-

tiation by their distinct biogenesis would be definite, but as this is not usually feasible, a

commonly used criterion is the vesicle size (Vlassov et al. 2012). Exosomes are secret-

ed by nearly all cells and can be found in most body fluids, such as blood, saliva, urine,

breast milk and semen (Caby et al. 2005; Poliakov et al. 2009; Lasser et al. 2011; Bo-

brie et al. 2011; Raj et al. 2012). They encapsulate mRNA, miRNA, siRNA, dsDNA,

proteins and other constituents within their bilayer coating (Valadi et al. 2007;

Wahlgren et al. 2012; Thakur et al. 2014). The inner composition of these vesicles de-

pends on the type of cells they originate from. Additionally, exosome cargo is selective-

ly sorted depending e.g. on the environment the cells are subjected to (Valadi et al.

2007; Ramteke et al. 2013; Martins et al. 2013). Cancerous cells exhibit different exo-

some composition compared to their non-cancerous counterparts and exosome amount

is often increased. The exosome research thus raises a possibility of a new detection

method to identify different cancer types at their early stage (Lu et al. 2009). Exosomes

are believed to function as message carriers, as they are proven to be able to fuse with

other cells and to deliver their cargo over a distance within body (Jung et al. 2009). Ex-

osomes contain membrane transport and fusion proteins that enable them to recognize

the target cells and to deliver their cargo. Internalization of exosomes by their target

cells initiates responses in these cells. For example, exosomes can activate PI3K and

ERK mediated signaling pathways in their target cells (Qu et al. 2009). They are shown

to promote cancer propagation and metastasizing and thus, are promising candidates as

targets for new cancer treatments (Peinado et al. 2012). Alternatively, they could be

used as targeted carriers for anticancer drugs.

Page 50: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

44

Prostate cancer is the most common cancer among male population and one of the most

common causes of death from cancer in men over 60 in western countries (Canadian

Cancer Statistics 2012; Ferlay et al. 2013; Siegel et al. 2013). Common practice for de-

tection of prostate cancer includes physical examination of the prostate, biopsy and de-

tection of prostate specific antigen (PSA) from the blood (Mäkinen et al. 2002; Hayes

and Barry 2014). None of these methods are conclusive for early detection of prostate

cancer, and exosomes could offer a possibility for a novel early biomarker. Prostate

cancer cell line LNCaP (Lymph Node Cancer of Prostate) is known to excrete exo-

somes also in vitro (Ramteke et al. 2013). Commonly used protocol to isolate exosomes

is separation by ultracentrifugation (Ramteke et al. 2013; Bijnsdorp et al. 2013; Liu et

al. 2014). It has been shown that SPR is capable to detect cell reactions to different

stimulus that cause cell morphology changes or intracellular events. Thus, it is expected

that the uptake of exosomes into prostate cancer cells provoke such responses, including

endocytosis and / or intracellular signaling. The use of SPR would offer a cheap and fast

method to investigate cell-exosome interactions. It would allow the detection of exo-

some internalization in real time and provide kinetic data of the interaction. Additional-

ly, SPR could offer a method to investigate if different cell types react to specific exo-

somes, or if specific cell type reacts to exosomes from different origin.

9 AIM OF THE STUDY

No studies on the internalization of exosomes by prostate cancer (LNCaP) cells have

been done previously. Aim of this study is to investigate whether these cells are able to

uptake exosomes isolated from different sources. Also, SPR as a new method for detec-

tion of exosome uptake is introduced. Polyethyleneimine (PEI)/DNA nanoparticles are

known to effectively penetrate into cells and SPR has been successfully used to detect

their cellular uptake (Silén 2013). Thus, PEI/DNA polyplexes are used as a positive

control. Exosomes excreted by LNCaP cells are separated, and exosomes from platelet

and prostate cells are introduced to a confluent monolayer of LNCaP and PC-3 (prostate

cancer cell line) cells to detect exosome internalization. The ability of LNCaP and PC-3

cells to adhere and form a confluent cell monolayer on a gold substrate is investigated

Page 51: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

45

without and with different cell adhesion promoters. Traditionally, peak angular position

(PAP) is evaluated in SPR based cell sensing. In this work, also peak minimum intensi-

ty (PMI) and TIR angle changes are examined to deepen the understanding of cellular

reactions to exosomes and PEI/DNA nanoparticles.

10 MATERIALS AND METHODS

10.1 Overview of the study

LNCaP cells were cultured for harvesting exosomes from collected growth medium.

Exosome separation was done by ultracentrifugation. Exosome protein concentration

was determined with protein assay and particle amount and size distribution with nano-

particle tracking analysis (NTA). LNCaP and PC-3 cells were then cultured on a gold-

coated sensor slide, with or without adhesion enhancers. The viability and confluency of

cells on a sensor were determined and platelet exosome, prostate exosome and

PEI/DNA polyplex interactions with cell monolayers were observed by SPR.

10.2 LNCaP and PC-3 cell culture

LNCaP cells (ATCC, Manassas, VA, USA) were cultured in Roswell Park Memorial

Institute (RPMI) 1640 medium (ATCC modification) (Gibco) supplemented with 0.4 %

(v/v) penicillin-streptomycin (Gibco) (final concentration 40 IU/ml penicillin and 40

µg/ml streptomycin) and 10 % (v/v) heat inactivated fetal bovine serum (FBS) (Gibco).

FBS was purified of endogenous exosomes and other vesicles by ultracentrifugation and

filtration. Serum was centrifuged for approximately 20 hours at 100 000 g at 4 °C and

then filtered with 0.22 µm Steritop filter unit (Millipore). PC-3 cells (ATCC) were cul-

tured in Ham's F-12 Nutrient Mixture (F-12y) (Gibco) supplemented with 0,4 % (v/v) of

penicillin-streptomycin and 10 % (v/v) of heat inactivated exosome free FBS. The com-

plete growth medium was filtered using 0.22 µm Steritop filter unit before use. Cells

were incubated at 37 °C under a humidified atmosphere of 5 % carbon dioxide (CO2) in

Page 52: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

46

75 to 175 cm2 cell culture flasks. Passage numbers ranged from 29 to 86 for LNCaP and

from 18 to 29 for PC-3 cells. Both cell lines were passaged twice per week using 0.25

% trypsin-ethylenediaminetetraacetic (EDTA) in PBS (Gibco) or TrypLE Express (Gib-

co). In addition, medium was changed once per week.

10.3 Exosome collection form LNCaP cells

To ensure good exosome yield, 20 ml and 45 ml of culture medium was used for 75 cm2

and 175 cm2 cell culture flasks, respectively. All medium used for LNCaP cell culture

was collected to 50 ml conical tubes and stored at -18 °C for further handling. Later,

frozen medium was thawed and centrifuged at 4 °C, 500 g for 10 min (Eppendorf 5810

R, Hamburg, Gesrmany) to remove cells and cell debris. This supernatant was further

centrifuged at 4 °C, 1200 g for 20 min, followed by centrifugation at 4 °C, 20 000 g for

60 min (Sorvall RC-5C Plus, Kendro Laboratory Products, Newtown, CT, USA) to re-

move apoptotic bodies and vesicles larger than exosomes. After this, supernatant was

filtered using 0.22 µm Steritop filter unit. The filtered supernatant was further processed

in two ways: 1) direct ultracentrifugation and pellet resuspension in phosphate buffered

saline (PBS) or 2) concentration before ultracentrifugation and storage as pellets. In the

first method, filtered supernatant was directly ultracentrifuged at 4 °C, 100 000 g for 60

min (Beckman Coulter Optima LE-80K, Brea, CA, USA). Supernatant was aspirated

carefully and exosome pellet was resuspended by shaking and pipetting repeatedly each

tube with 200 µl of cold PBS. These exosome samples were stored at -18 °C. Later,

samples were pooled and measured for their exosome protein content (described in sec-

tion 10.4.2). The samples were divided to aliquots of 6 µg protein (88 µl) and stored at -

80 °C. In the second method, the protein amount of filtered supernatant was determined

and 19-22 ml of supernatant was concentrated to maximum volume of 1 ml using

Amicon Ultra-15 centrifugal filter device with 100 000 nominal molecular weight limit

(NMWL) (Millipore). This concentrate was then ultracentrifuged in 1.5 ml centrifugal

tubes at 4 °C, 100 000 g for 60 min. Tubes were aspirated carefully and dry exosome

pellet samples were stored at -80 °C.

Page 53: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

47

10.4 Protein amount determination

Exosomes in PBS and pellets were tested for their protein content in order to evaluate

the final exosome yield. For the PBS samples one aliquot (88 µl) was used, whereas

pellet samples were first resuspended into 100 µl of PBS by repeated pipetting and

shaking. When determining protein concentration from e.g. filtered supernatant, 100 µl

was taken for sample.

10.4.1 Sample washing

In order to remove non-exosome proteins, the samples were washed using Amicon Ul-

tra-4 centrifugal filter device with 100K NMWL (Millipore). 400 µl of PBS (without

calcium / magnesium) was added to samples and centrifuged at 4 °C, 3130 g for 15-20

min. Rinsing was repeated twice. To ensure the removal of larger protein constituents,

the remaining filtrate was supplemented with 10 % (v/v) 10X trypsin-EDTA (Gibco)

and incubated for 30 min at 37 °C followed by rinsing with 400 µl of PBS three times.

Last filtrate was suspended to final volume of 600 µl of PBS.

10.4.2 Protein assay

Micro BCA protein assay kit from Thermo scientific was used to determine protein

amount of the exosomes. Washed samples were put to 95 °C for 10 minutes to break

down exosomes and to release their protein content. 1:10 and 1:100 dilutions of the

samples were made in PBS. 150 µl of non-diluted- and diluted samples and standards of

known protein amount were pipetted on a flat bottom 96 well plate. 150 µl of micro

BCA working reagent was added to each well and shaken. Plate was sealed with para-

film and incubated at 37 °C for 2 hours. The plate was cooled to room temperature and

absorbance was measured at 562 nm wavelength using Varioskan flash multiplate read-

er (Thermo scientific, Waltham, MA, USA).

Page 54: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

48

10.5 Exosome particle size distribution

Particle size distribution of exosomes collected from LNCaP cells was measured by na-

noparticle tracking analysis. Pellet samples were resuspended with PBS to a final vol-

ume of 300 µl. Dilution series of 1:10 to 1:100 000 were prepared and NTA analysis

was performed using the NanoSight LM10 (Malvern, Worcestershire, United Kingdom)

nanoparticle tracking analysis instrument.

10.6 Gold substrate surface modifications

Gold-coated SPR sensor slides (12 x 20 mm) were obtained from Bionavis Ltd (Tampe-

re, Finland). Sensors were cleaned prior to their use by boiling them for 5-10 minutes in

a solution containing one part of 30 % ammonium hydroxide solution (Sigma-Aldrich),

one part of 30 % hydrogen peroxide solution (Sigma-Aldrich) and five parts of Milli-Q

water. After boiling, sensors were rinsed with Milli-Q water, dried with airflow and

sterilized by autoclaving. Finally, sensor slide surface was treated with three different

cell adherence enhancers: rat tail collagen I (Cultrex), human plasma fibronectin (HFN,

Millipore) and CELLstart (CTS, Gibco).

Rat tail collagen I (3 mg/ml) and Human fibronectin (1 mg/ml) were diluted to desired

concentrations using 0.02 M acetic acid and PBS, respectively. CELLstart was diluted

with PBS at a ratio of 1:50 according to manufacturer’s guidelines. 2 ml of collagen and

fibronectin and 0.7 ml of CELLstart were pipetted in an 8.8 cm2 culture dish with a gold

sensor. Collagen and fibronectin treated sensors were incubated for 1 hour at room tem-

perature and CELLstart treated sensor was incubated at 37 °C, 5 % CO2 for 2 hours, af-

ter which solutions were aspirated. Collagen and fibronectin treated sensors were rinsed

with PBS. All sensors were seeded with the cells immediately after the treatment. For

collagen, dilutions resulting in a culture dish coverage of 4.5-5.5 µg/cm2 were tested

and for fibronecting 4.5-10 µg/cm2.

Page 55: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

49

10.7 Immobilization of cells on gold substrate

Cultured cells were detached using trypsin-EDTA or TrypLE Express and re-suspended

with complete growth medium. Cell counting and viability testing was done using Try-

pan blue exclusion. Trypan Blue Stain 0.4 % (Gibco) was added into cell solution at 1:1

ratio (v/v) and cell count and viability was measured using Cedex XS cell counter

(Roche Diagnostics, Basel, Switzerland). Medium was then added to adjust the cell

concentration.

Sterilized, surface treated and non-treated gold sensors were placed on a culture dish

with a surface area of approximately 8.8 cm2. Desired amount of resuspended cells and

pure medium was added to final volume of 2 ml and plates were incubated at 37 °C, 5

% CO2. Cell attachment and confluence on gold sensor was observed after 24 to 96

hours with microscope. At the end of the incubation, when growth on gold sensor was

near confluent, cell viability was assessed using Trypan blue exclusion method. Trypan

Blue Stain 0.4 % and medium was added into culture dish at 1:1 ratio (v/v) and incubat-

ed at 37 °C, 5 % CO2 for 2-3 minutes. Viability was evaluated by eye; Dead cells take

up dye making them readily distinguishable, while living cells do not.

10.8 Sample preparation for surface plasmon resonance

The volume of the flow cell chamber is 100 µl and the tubing adds roughly another 100

µl of dead volume. Minimum sample volume of 300 µl was used to ensure sufficient

sample amount to fill the entire measurement chamber and to avoid injection of air.

10.8.1 Exosome samples

Thrombin-collagen activated platelet exosome samples were obtained from Department

of Biosciences, Biochemistry and Biotechnology, University of Helsinki. The protein

content of these exosome pellet samples was 6 µg. For pellet resuspension, 50 µl of se-

rum free medium was added and mixed vigorously by pipetting several times. Medium

Page 56: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

50

was then added to final volume of 300 µl, resulting in final sample concentration of 20

µg/ml.

Prostate exosome samples collected from LNCaP cells containing 6 µg of protein in 88

µl of PBS were diluted with serum free medium to final volume of 300 µl. Dry exosome

pellet samples from LNCaP cells were first resuspended with 50 µl of medium, mixing

vigorously by pipetting several times, followed by addition of medium to final volume

of 300 µl.

10.8.2 PEI/DNA polyplex samples

Plasmid DNA (pDNA) coding β-galactosidase (pCMVβ) and luciferase (pCLuc4) were

received as a gift from University of California, San Francisco, CA, USA. Amplifica-

tion, isolation and purity analysis were performed by Martina Hanzlíková and others at

University of Helsinki (Hanzlíková et al. 2011).

A stock solutions of 1 mg/ml pDNA in Tris-EDTA (TE) buffer solution (Sigma-

Aldrich) and 1 mg/ml branched polyethyleneimine (PEI) with mean molecular weight

of 25 kDa (Sigma-Aldrich) in water at pH 7 were prepared. Into 4 µl of DNA and 6.4 µl

of PEI stock solutions, MES-HEPES buffer with pH 7.4 (Sigma-Aldrich) was then add-

ed to dilute and adjust the pH. Buffered PEI solution was then added into buffered DNA

solution and incubated for 20 minutes at room temperature. This results in PEI/DNA

polyplexes with an N/P ratio of 12 (the amount of positively charged constituents in

moles is 12 times that of negatively charged). After incubation 310 µl of MES-HEPES

buffer and 1600 µl of serum free medium were added. Total volume of the final

PEI/DNA polyplex sample was 2 ml with a PEI/DNA concentration of 5.2 µg/ml.

10.9 Surface plasmon resonance experiments

Experiments were performed using an SPR Navi 200 instrument (Bionavis Ltd, Tampe-

re, Finland). Prior to testing, the flow cell and tubings were rinsed with 70 % ethanol

Page 57: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

51

and Milli-Q water, and the flow cell chamber was warmed to 37 °C. To ensure that no

contaminants from serum affect the experiment, serum free medium was used in all ex-

periments. For samples containing PBS in addition to growth medium, a solution with

29 % (v/v) of PBS in serum free growth medium was prepared to correspond the

amount of PBS in the samples. All samples and medium were preheated to 37 °C.

Sensor slides with cells were inserted to the slide holder and the glass side of the sensor

was cleaned with 70 % ethanol to remove any impurities that could affect optical prop-

erties of the sensor slide. The slide holder with the sensor slide was then quickly insert-

ed into the SPR device and medium was injected. After initial injection the cells were

allowed to adjust for 15-20 minutes, after which the sample was injected. All experi-

ments were carried out by batch method i.e. there is no flow between injections. Injec-

tions were done manually using a syringe. Typical observation time after sample injec-

tion was approximately 30 minutes, after which sample was removed by injecting me-

dium, and cells were observed for another 20-30 min. Experiment was ended with re-

moval of the medium, and sensor slide was quickly placed in a cell culture dish with

complete growth medium. Immediately after the experiment, the sensor slide was ob-

served under microscope for cell monolayer integrity. Also cell viability was evaluated

with Trypan blue exclusion method as described earlier in section 10.7.

Before each experiment, an initial scan comprising the full SPR angular spectrum of 40-

78 ° was performed. Angular scan spectrum of 57-75 ° was then used for all experi-

ments as this range is sufficient to cover the whole refractive spectrum in our liquid

samples. This angular spectrum was scanned every 2.24 seconds. Scanning was per-

formed using two wavelengths simultaneously, 783 and 668 nm. All data presented is

obtained form wavelength of 783 nm. As the evanescent field depth (δz) of surface

plasmons is dependent on the wavelength of incident light, longer wavelength extends

deeper into the sample medium. Thus, by using wavelength of 783 nm, we are able to

see deeper into the cell. In addition to peak angular position, peak minimum intensity

and total internal reflection (TIR) area were evaluated. Using all these parameters, we

are able to gain deeper understanding of cellular reactions to test compounds.

Page 58: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

52

11 RESULTS AND DISCUSSION

11.1 Exosome yield, particle count and size distribution

As detecting exosomes directly is complicated, the exosome amount can be expressed

in relation to protein content of exosomes. From Micro BCA assay, the exosome yield

was determined to be an average of 2.8–14 µg from 6-43 × 10 cells. The protein

amount in pelleted sample aliquots was found to be from 2.7 to 14 µg per pellet, result-

ing in 9-47 µg/ml in 300 µl final sample to be used in SPR experimentation. As protein

assay indicates, exosome samples prepared by ultracentrifugation can vary greatly in

their exosome amount. For more homogenous samples, further optimization of separa-

tion process would be required.

NTA uses laser light source to illuminate nano sized particles and determines the count

and size distribution by tracking Brownian motion of the particles. It is capable of dif-

ferentiating particles with diameter of 10-2000 nm. The particle size distribution gives

an approximation on success of separation by ultracentrifugation. However, this method

cannot discriminate exosomes from other vesicles with the same size, and samples can

only be regarded as exosome enriched rather than pure exosome samples. The NTA as-

say showed that the final prostate exosome sample aliquots had an average particle size

of 154 nm with standard deviation of 102 nm (Figure 27). This mean particle size is at

the upper limit of expected exosome size. However, due to NTA assay characteristics

and exosome aggregation contributing to measured mean particle size, the size distribu-

tion falls well within the exosome size range (Sokolova et al. 2011; Ramteke et al.

2013). The particle count was 6.1 × 10 particles ml⁄ in 1:10 diluted sample, i.e.

6.1 × 10 particles ml⁄ in undiluted sample.

Page 59: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

53

Figure 27. Average particle amount and size distribution in prostate exosome samples. Y-axis shows number of particles (particles × 10 ml⁄ ) and x-axis the particle size (nm). Black area indicates the standard error of the mean.

11.2 Cell immobilization on gold sensor

To ensure repeatability of SPR experiments, cells have to attach on the gold surface of

the SPR sensor slide and form a tight monolayer. Bare spots on the sensor can cause

inconsistent signals on sensograms. On the other hand, cells stacked in more than one

layer might obscure the detection of cell-particle interactions. Also, cells have to with-

stand the strain inflicted upon them during the SPR measurement, as the medium and

sample injecting causes a flow of liquid inside the flow cell chamber.

11.2.1 LNCaP cells

First, cells were seeded on top of gold sensors using cell densities of 7 × 10 , 1 × 10 and 1.5 × 10 cells cm⁄ . The concentrations were chosen based on the cell line

supplier (ATCC) recommendations and previous publications (Hiragun et al. 2012). Af-

Page 60: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

54

ter 24 hours, the two lower seeding densities showed cells proliferating and attaching on

the sensor, but not covering the whole sensor surface. The highest seeding density of

1.5 × 10 cells cm⁄ on the other hand covered the whole sensor almost completely

after 24 hours (Figure 28).

Figure 28. Light microscopy images of LNCaP cells growing on gold sensor slide. a) - c) show the sensors after 24 hours of seeding, with seeding densities of 7 × 10 , 1 × 10 and 1.5 × 10 cells cm⁄ , respectively. d) - f) represent sensors after 48 hours with seeding densities of 7 × 10 , 1 × 10 and 1.5 × 10 cells cm⁄ , respec-tively. g) shows a sensor with 1 × 10 seeded cells cm⁄ after 96 hours and h) 1.5 × 10 seeded cells cm⁄ after 72 hours. The scale bar in all images is 200 µm.

Page 61: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

55

After 48 hours, all seeding densities showed that cells had proliferated to cover the sen-

sors completely or almost completely. The highest seeding density of 1.5 × 10 cells cm⁄ was so full that minor cell clumping begun to emerge. If the cells are

incubating further for 72–96 hours, they grow past the confluency and start to stack on

top of each other and form clumps (Figure 28 g) and h)). Trypan blue exclusion test ver-

ified that cells are alive on the sensor slides regardless of seeding density. However, the

assessment of the most suitable seeding density is complicated. The lowest amount of

cells in most cases form an adequate monolayer within 48 hours. The highest cell seed-

ing density provides confluent monolayer more reliably, but after 48 hours the risk of

clumping is evident. Furthermore, higher passage number of the cells increased the cell

proliferation on the sensors and lower seeding density was required for cells at higher

passage number.

To further asses if the sensor with highest seeding density of 1.5 × 10 cells cm⁄

could be used already after 24 hours incubation, an SPR experiment was conducted us-

ing only growth medium as injected sample. The result shows that even if the cells are

distributed to the whole sensor area, the strength of attachment is not sufficient and cells

are detached during the SPR run (Figure 29).

Next, surface modified sensor was tested to see if cell adhesion promoting agent could

expedite cell adhesion to the sensor surface. Lower seeding density of 7 × 10 cells cm⁄ was used, as it was expected that adhesion protein could decrease the

amount of cells required to cover the whole sensor area. Indeed, a sensor treated with

5.5 µg/cm2 collagen I showed that despite the lower seeding density, sensor was com-

pletely confluent 24 hours after seeding. Yet, from figure 29 it can be seen that even

with the collagen treated sensor, the attachment of cells was still not strong enough and

cells detached during the SPR run.

Page 62: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

56

Figure 29. Light microscopy images of LNCaP cells before and after SPR experimenta-tion on adhesion protein treated and non-treated sensors with 20-24 hours incubation. a) shows the sensor after 24 hours incubation with no adhesion protein with cell seeding density of 1.5 × 10 cells cm⁄ . b) shows sensor after 20 hours incubation, treated with 5.5 µg/cm2 collagen I with seeding density of 7 × 10 cells cm⁄ . c) and d) show sensors a) and b) after SPR experiment, respectively. The scale bar in all images is 200 µm.

In addition to collagen, two other adhesion proteins, fibronectin and CELLstart, were

also experimented to estimate their effectiveness on LNCaP cell adhesion. Collagen,

fibronectin and CELLstart were used in different concentrations and seeding densities

(Table 2). Higher seeding densities were not experimented, as it was evident that lower

cell concentrations were sufficient for the formation of confluent monolayers when us-

ing adhesion promoters.

Page 63: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

57

Table 2. Cell and adhesion protein concentrations tested on gold sensor surface.

Cell concentration (cells/cm2)

Tested without adhesion pro-

moter

Fibronectin (µg/cm2)

Rat tail colla-gen I (µg/cm2)

Cellstart solu-tion (µl/cm2)

5 × 10 no 10 - -

7 × 10 yes 4.5 10

4.5 5.5 80

1 × 10 yes - - -

1.5 × 10 yes - - -

As the experiments with collagen already indicated, these adhesion enhancers do not

provide adequate attachment after 24 hours incubation. Thus, all other adhesion pro-

moter tests were conducted using 48 hours incubation time (Figure 30). According to

the results, after 48 hours of incubation, cells remained on both untreated and surface

modified sensors after the SPR run. Seeding density had no effect on how strong cells

attach on sensors and both seeding densities of 7 × 10 and 1.5 × 10 cells cm⁄

sustained a nearly uniform cell monolayer under the stress of the SPR run. The medium

flow caused by injection during the assay always causes some cell detachment, espe-

cially at the vicinity of the flow cell inlet and outlet. This is most likely due to the turbu-

lence within the flow cell chamber at the site where medium flows in and out, and it can

be seen as cell monolayer deterioration in these areas. Sensors with adhesion enhancers

showed small improvement in this; all sensors treated with any of the adhesion promot-

ers showed more consistent cell monolayer throughout the flow cell chamber area.

However, the cell monolayer deterioration around the edges of the flow cell chamber

should not pose a problem. This is because the site of measurement area is small and is

located at the very bottom of the flow cell chamber area. Also, SPR parameters, such as

TIR area shape, provide information if the measurement area has bare gold surface ex-

posed. Adhesion promoter treated or untreated sensors showed no differences in SPR

responses for test compounds used (data not shown). Thus, the results are comparable

regardless of sensor treatment. This conclusion is supported by previous studies where

adhesion promoters were used (Wang et al. 2012a; Chabot et al. 2013).

Page 64: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

58

Figure 30. Light microscopy images of LNCaP cells before and after SPR experiment on adhesion protein treated and non-treated sensors with 48 hours incubation. a)-e) show sensors before and f)–j) after SPR run. The conditions were as follows: a), b), f) and g): no adhesion enhancer, c) and h): 4.5 µg/cm2 of collagen I, d) and i): 10 µg/cm2 of fibronectin and e) and j): 80 µl/cm2 of CELLstart solution. Seeding density for b) and g) was 1.5 × 10 cells cm⁄ and for all the rest 7 × 10 cells cm⁄ . The scale bar in all images is 200 µm.

Page 65: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

59

The results from experiments with LNCAP cell immobilization on gold sensor can be

summarized in three main conclusions: 1) Cell seeding density is somewhat flexible for

LNCaP. Both the lower and the higher seeding densities result in coherent cell mono-

layer. Nevertheless, cell proliferation is always variable and may vary even when using

same seeding density. In addition, it was evident that the passage number of the cells

plays a role in how fast they proliferate. With larger passage number the cell monolayer

formation is accelerated. Thus, for the SPR experiments the cell seeding density varied

and both lower and higher densities were used. 2) 24 hours is not enough for cells to

attach on gold surface strong enough to endure the stress under SPR run. Minimum of

48 hours incubation is needed. 3) Adhesion promoters do not shorten the time needed

for cells to adhere to gold surface. Instead, they do help cell monolayer to remain more

consistent on the sensor surface during the SPR measurement. In addition, they do not

affect SPR responses, and results are comparable whether adhesion promoters are used

or not.

11.2.2 PC-3 cells

PC-3 cell line showed very similar behavior than LNCaP cells when incubating on gold

substrate. Proliferation was good and cells attached strong enough to withstand the

stress under the SPR run. Seeding density had more pronounced effect on monolayer

character than with LNCaP. A seeding density of 1 × 10 cells cm⁄ seemed to pro-

duce the most consistent monolayer repeatedly, whereas 1 × 10 cells cm⁄ often

showed bare spots. Cell stacking started to appear with seeding density of 1.5 × 10 cells cm⁄ . As with LNCaP, higher cell line passage number increased the prolif-

eration rate, and confluency on sensor slides was reached faster. In addition, the degree

of cell stacking was sensitive to cell passage number, and earlier passages did not show

strong clumping with the higher seeding density. 48 hours of incubation was needed for

cells to gain sufficient attachment. Trypan blue exclusion assay verified viability of PC-

3 cells on gold sensor after 48 hours and also after the SPR run. No adhesion promoters

were tested. Depending on cell passage, seeding densities of 1 × 10 and 1.5 × 10 cells cm⁄ were used for SPR experiments. Unfortunately, due to a contamination

Page 66: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

60

of this cell line, we were unable to undergo some of the planned SPR experiments with

these cells, namely, none of the experiments with exosomes were conducted.

11.3 Surface plasmon resonance experiments

11.3.1 Initial scan

The integrity of cell monolayer on gold sensor was evaluated by examining each sensor

under microscope before experimentation. Often, the growth was not complete to the

very edges of sensor slide. However, this should not pose a problem as the flow cell

chamber positions at the bottom middle of the sensor slide. Also, SPR Navi 200 uses

multiple lasers, others positioned at the top and others at the bottom of the flow cell

chamber. In our experiments, we utilized the bottom lasers and sensor slides were

placed accordingly. If cell monolayer was not completely confluent, slides were placed

in such a way that the confluent part of the monolayer was at the bottom. Further evalu-

ation of the cell monolayer integrity was then made by observing the full SPR curve be-

fore each test. Figure 31 shows a full SPR curve for a sensor slide without cells and

with LNCaP monolayer. When cell monolayer is present, the TIR region shows a more

round shape and is shifted to higher angle, from around 61 ° to 63 ° and to lower inten-

sity, from around 0.81 to 0.78. Also PAP shifts from 66 ° to 67.5 ° and PMI increases

from 0.05 to 0.1. These chances are in accordance with experiments done by Yashunsky

et al. (2010b) and Viitala et al. (2013). Yashunsky et al. monitored cell monolayer for-

mation concurrently with optical microscopy and SPR. Viitala et al. simulated a full

SPR curve of a thick sample layer on the sensor surface, corresponding to SPR curve

produced by a cell monolayer. In both experiments the cell monolayer caused the PAP

to shift to higher angle and PMI to increase. In addition, Viitala et al. showed similar

TIR region changes as observed here.

Page 67: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

61

Figure 31. A full SPR curve of pure gold sensor (blue dashed line) and sensor with LNCaP monolayer (solid red line). Graph shows a typical shape alteration of SPR curve when cell monolayer is present. Peak minimum intensity and angular position increase. TIR region shifts to lower intensity and to higher angle. Also, typical TIR region round-ing is noticeable.

After initial medium injection, PAP shows some shifting. This shift can be an increase

or decrease in PAP while the cells react to the environmental change. Within 10 to 15

minutes, we could see the chance in PAP settling to either constant value or constant

slope. In the results presented here, this initial shifting of sensograms is not included to

clarify the actual SPR responses to samples. Also, because several sources of variation

exist, including differences in the concentration of exosome samples, cell passage num-

ber and cell occupancy on sensor, individual SPR sensograms vary in observed re-

sponse. Especially, in some experiments with exosome samples, the background signal

obscures the actual response almost completely. For these reasons, only representative

data is presented.

11.3.2 Peak angular position and total internal reflection area angle

As cells are known to uptake PEI/DNA polyplexes, they were used as a positive control.

For negative control, a pure medium was used. The injection of medium without sample

Page 68: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

62

causes a sharp increase in PAP, followed by a quick return to initial value as seen on

figure 32 a). The same is observed also with PEI/DNA sample, after second injection of

plain medium that ends PEI/DNA stimulation in figure 32 b). With the first injection

with PEI/DNA sample, this transient peak is most likely obscured by the large shift in

PAP caused by PEI/DNA itself. Other research groups also observed these transient

peaks with other cell lines when cells were forced to changes in their cell size (Robelek

and Wegener 2010; Baumgarten and Robelek 2011). These peaks were later abolished

by the use of long-range surface plasmons (LRSP) that probes up to three times deeper

into the sample layer (Vala et al. 2013). Thus, it was concluded that the short penetra-

tion depth of surface plasmons is sensitive to local changes in refractive index that do

not accurately represent the cell as a whole. Wang et al. (2012b) attributed these local

changes as vertical cell membrane movement, when cells were stimulated with non-

isotonic medium. To evaluate the actual cell responses to the used stimulus, these tran-

sient peaks should be ignored. It is safe to assume that also the transient peaks observed

in our experiments are of similar origin; medium injection causing minor local changes

within the evanescent field e.g. cell monolayer pushed towards the sensor. By using

method such as LRSP that penetrates deeper into the sample layer, these peaks would

be rendered nonexistent.

Figure 32. Change in PAP with a) pure medium sample and b) PEI/DNA polyplex sam-ple representing negative and positive controls, respectively. Solid arrow indicates in-jection of sample and dashed arrow marks injection of medium. y-axis shows change of PAP in degrees and x-axis time in minutes.

Page 69: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

63

When neglecting these transient peaks, the results show that medium sample does not

induce any shift in PAP. For PEI/DNA on the other hand, the PAP shift is formidable, a

0.9 ° increase. After washing the sample, no changes in PAP occur. When inside the

cells, PEI/DNA particle trafficking includes movement toward inner parts of the cells

and fusing with lysosomes (Godbey et al. 1999). In the time course of SPR experiment,

no particle recycling out of the cells could occur. Also, attachment/detachment from the

cell surface or delayed internalization should show changes later in the SPR signal.

Thus, the increase in PAP seems to be due to PEI/DNA nanoparticle endocytosis by

LNCaP cells. These control experiments with nanoparticles and pure medium sample

were repeated with PC-3 cell line. The experiments showed similar responses as with

LNCaP cells (Appendix figure A1).

In the work done by Viitala et al. (2013), where the full SPR angular spectra of a thick

sample layer were simulated, the sample layer properties were set to produce SPR sig-

nals that a confluent cell monolayer produces. They divided the cell monolayer into two

theoretical regions: a lower part within the evanescent field and an upper part outside

the reach of evanescent field. Changes at different regions of the cell monolayer were

simulated for their effect on SPR signals. Also, MDCK cell monolayer was used to

compare these simulated responses to experimental signals. Total internal reflection

(TIR) area has not been utilized earlier in cell sensing. However, Viitala et al. showed

that this region within the SPR curve could further elucidate cellular responses. Accord-

ing to the simulation, the angle of the TIR region reflects refractive index changes in

regions outside the evanescent field within the cell monolayer. Indeed, in their experi-

ments with MDCK cells, TIR angle correlated with PAP angle for changes in refractive

index within the cell monolayer. The instrumentation used in our experiments does not

support direct data acquisition of TIR angle. However, from full SPR curve it is possi-

ble to manually follow intensity changes at the quasi-linear slope of the TIR area. The

fixed angle intensity changes obtained this way translate into TIR region angle changes,

i.e. increase in intensity when TIR angle decreases and vice versa. This is a similar

method as when following peak angular changes using fixed angle scan. The TIR area

angle sensograms acquired by this method are presented as a relative change and cor-

rected to show positive values for increasing TIR angle and vice versa. From figure 33,

Page 70: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

64

it can be seen that TIR angle alterations with LNCaP cells for medium sample a) and

PEI/DNA sample b) correlate perfectly with PAP sensograms from figure 32. This fur-

ther confirms the fact that the observed responses actually originate from mass accumu-

lation within the cell monolayer.

Figure 33. TIR angle sensograms for a) pure medium sample b) PEI/DNA polyplex sample. Solid arrow indicates the injection of sample and dashed arrow injection of pure medium. y-axis scale shows relative change between samples. x-axis shows time in minutes.

Next, platelet and prostate exosomes were tested on LNCaP cells. After exosome injec-

tion, an increase in refractive index is seen (Figure 34). This increase of 0.1 ° is much

smaller, compared to what is seen with the PEI/DNA sample, yet the response is clear

when compared to the negative control sample. For the platelet exosome sample and the

medium injections, similar initial transient peaks as in the control experiments are seen.

For prostate exosome sample, this transient peak seems to be obscured by stronger shift

in PAP caused by the sample itself, as was with PEI/DNA sample. As previously ex-

plained, these peaks are ignored. Moreover, for platelet exosomes, an initial decrease in

PAP is seen that is not observed with prostate exosomes. A similar phenomenon is seen

with cells rearranging their actin cytoskeleton. Cytoskeleton movement towards basal

cell membrane increases the RI, but this positive change in PAP is obscured by a strong

negative signal caused by cellular contraction (Cuerrier et al. 2008; Chabot et al. 2013).

As cells later regain their morphology and spread, the PAP increase caused by cytoskel-

Page 71: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

65

eton rearrangement becomes visible. It seems that LNCaP cells react to platelet exo-

somes in a similar manner. The initial decrease in PAP caused by cellular contraction is

quickly shifted to PAP increase as exosomes move into cells and cells regain their orig-

inal morphology. LNCaP cell reaction to prostate exosomes seems to differ, as the only

response observed is the increase in PAP, resulting from either cellular spreading or ex-

osome accumulation within cells or both. Reason for this difference in the responses is

unknown.

Figure 34. Change in PAP with a) LNCaP exosomes and b) platelet exosomes. Solid arrow indicates the injection of sample and dashed arrow injection of pure medium. y-axis shows change of PAP in degrees and x-axis time in minutes.

Differences in SPR response between these exosomes emerge also after rinsing the

sample with plain medium on the second injection. When rinsing prostate exosomes,

PAP returns to its original value, whereas for platelet exosomes no change in PAP is

seen, i.e. the angular position is shifted to higher values permanently. This suggests that

prostate exosomes exit the cells when pure medium is added, whereas platelet exosomes

are retained within the cells. This is reasonable, as prostate exosomes originate from

these same cells. Thus, these cells have the inherit capability to excrete these exosomes.

On the contrary, platelet exosomes are not naturally present in these cells, and conse-

quently are not expected to excrete them.

Page 72: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

66

The TIR angle sensogram for prostate exosomes shows the same transition phases as

the PAP sensogram (Figure 35 a)). For the platelet exosomes, the TIR angle is some-

what more difficult to interpret, but the same characteristics in responses as seen in PAP

sensogram, are present (Figure 35 b)). Upon exosome injection, a transient peak is ob-

served, followed by a decrease in the TIR angle. Then, continuous increase of TIR angle

follows. It seems that the platelet TIR sensogram after 20 minutes does not correlate

with the platelet PAP sensogram. However, this difference is a result of data handling

and manual acquisition of the TIR angle sensogram. When observing the TIR sen-

sogram from 20 minutes onwards separately, it is obvious that medium injection does

not have any effect on the sensogram and thus, it corresponds to the platelet PAP sen-

sogram. Exosome interactions on PC-3 cells were not experimented because of contam-

ination of the cell line.

Figure 35. TIR angle changes for a) prostate exosomes and b) platelet exosomes. Solid arrow indicates the injection of sample and dashed arrow injection of pure medium. y-axis scale shows relative change between samples. x-axis shows time in minutes.

11.3.3 Peak minimum intensity

To further evaluate cell monolayer response to exosomes, the PMI changes were evalu-

ated. The plain medium injections show very little response in PMI (Figure 36 a)). The

PEI/DNA sample, however, shows a considerable decrease in reflected light intensity

Page 73: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

67

(Figure 36 b)). This PMI change is opposite to the change in PAP sensogram of

PEI/DNA (Figure 32).

Figure 36. PMI change for a) pure medium sample and b) PEI/DNA polyplex sample. Solid arrow indicates injection of sample and dashed arrow marks injection of medium. y-axis shows the change in reflected light intensity and x-axis time in minutes.

Interpreting the intensity changes is challenging as very little effort has been made pre-

viously to understand these changes in SPR. However, the decrease in reflected light

intensity in this case is surprising, as according to current knowledge, the intensity is

expected to be parallel to PAP: PMI should increase as PAP increases and vice versa.

The intensity of reflected light depends on the losses of light at the sample layer due to

light scattering and/or absorption. In other words, the more light is scattered on the sur-

face, the less surface plasmons are generated and consequently, reflected light intensity

increases. Cell spreading on a sensor surface increases the mass at the evanescent field

and thus, the PAP and PMI increase (Yashunsky et al. 2010b). Accumulation of drug

into cells, causing cell layer spreading and mass redistribution in cells, is also expected

to result in an increase in both PAP and PMI (Viitala et al. 2013). On the contrary, cell

contraction and mass redistribution causes decrease in both sensograms. Cell volume

changes by fluid movement also cause corresponding PAP and PMI changes. When flu-

id flows out of cells, intracellular water content decreases, while concentration of intra-

cellular structures increases. Lowered water content leads to an increase in refractive

index and consequently an increase in PAP. The increased concentration of cellular

Page 74: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

68

structures increases the amount of light scattering surfaces causing PMI to increase

(Vala et al. 2013). When cells grow on the sensor surface to form a continuous mono-

layer, the cell occupancy on the sensor increases and thus PAP increases (Yashunsky et

al. 2010b). In this case, also PMI increases, because the surface area of cell membrane-

medium interfaces increase. However, when cells have reached a near confluent mono-

layer, PMI starts to decrease. This is because as the cell membrane monolayer on the

surface becomes more unified, the less light scattering interfaces exist. On the contrary,

PAP continues to increase, as long as more cellular content occupies the surface. Yash-

unsky et al. (2012b) also demonstrated that opposite PAP and PMI changes for conflu-

ent cell monolayer are possible. Chlorpromazine treatment causes corrugation (rippling)

of the basal cell membrane that leads to increased water amount within the evanescent

field. Increased water content within the sensing area causes PAP to decrease, while the

corrugated cell membrane increases the light scattering surface area and consequently

increases PMI. For a confluent cell monolayer, PMI can increase also if the cell mono-

layer integrity is disrupted (Yashunsky et al. 2010b). To understand cell reactions to

stimulants, differentiation between single cell and cell monolayer contribution to the

SPR signals must be made. For example, the increase in intracellular organelle concen-

tration, as cell water content is reduced, does not affect the cell monolayer occupancy

on the sensor, and the increase in both PAP and PMI are the result of increased concen-

tration of intracellular components (organelles) (Vala et al. 2013). When a confluent

cell monolayer is stimulated with drugs, PAP and PMI changes can be the result of both

intracellular events and cell monolayer changes resulting from accumulation of drugs

within the cells. The increase in PAP and PMI is a result of intracellular mass redistri-

bution towards the basal membrane and cell spreading, while decrease in PAP and PMI

results from intracellular mass redistribution away from the basal membrane and cell

contraction (Viitala et al. 2013). The intracellular mass redistribution can be thought as

increasing / decreasing water content locally within the cell. The reason for this is that

the SPR evanescent field senses only the very bottom of the cells. Local movement of

intracellular components, such as cytoskeleton rearrangement towards or away from the

basal membrane, is sensed by the evanescent field as an increase / decrease of cytoskel-

eton “concentration” at lower part of the cell.

Page 75: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

69

Explaining the reactions of LNCaP cells to PEI/DNA in not straightforward. While it is

evident that the increase in PAP is a consequence of cells internalizing these nanoparti-

cles, the SPR responses are difficult to interpret: as particles accumulate into the cells,

neither mass redistribution towards basal membrane and cell spreading, or mass redis-

tribution away from basal membrane and cell contraction can explain the opposite re-

sponses of PAP and PMI. The decrease in PMI could indicate a slight contraction of

cells, accompanied by mass redistribution away from evanescent field. However, in this

case, PAP should also decrease (Viitala et al. 2013). The increase of PAP indicates cell

spreading and/or mass redistribution towards basal membrane. In this situation, an in-

crease in PMI should occur. However, PAP increase along with PMI decrease can occur

if basal membrane of cell monolayer becomes more unified, a change opposite to the

membrane corrugation described by Yahsunsky et al. (2012b). Here, water content

within the evanescent field decreases, while at the same time less cell membrane-

medium interface is present. Considering all these aspects, it seems that the opposite

PAP and PMI signals are not caused by individual cell reactions only. It is likely that

these SPR responses are a combined result of PEI/DNA accumulation within the cells

and bulk changes within the cell monolayer. Mass redistribution and cell morphology

changes might occur, but changes in the cell monolayer can overcome these signals.

The intensity changes caused by exosome samples indicate similar responses to those of

PEI/DNA (Figure 37). For both, prostate and platelet exosomes, a decrease in reflected

light intensity can be seen. The decrease is more obvious for prostate exosomes (Figure

37 a)), while for platelet exosomes the signal is more complex (Figure 37 b)). With

platelet exosomes, after the initial drop in PMI, a continuous decrease in PMI remains.

No change in PMI is seen for either exosome samples, when rinsing with plain medium.

For platelet exosomes this is expected, as there is no change in PAP. The continuous

decrease in PMI for platelet exosomes is considered to correlate with the platelet PAP

sensogram, as already described for platelet exosome TIR angle results. For the prostate

exosome sample, lack of PMI change with plain medium injection is somewhat surpris-

ing. This is because the PAP returns to same position as before exosome stimulation.

This could mean that the initial decrease in PMI does not originate from intracellular

responses, but from bulk cell monolayer changes. Alternatively, some other response

Page 76: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

70

might obscure the PMI signal return when plain medium is injected. Finally, it is also

possible that the decrease in PAP does not originate from cell-exosome interactions.

Figure 37. PMI change for a) prostate exosomes and b) platelet exosomes. Solid arrow indicates injection of sample and dashed arrow marks injection of medium. y-axis shows the change in reflected light intensity and x-axis time in minutes.

Monitoring the TIR area intensity changes can be used to confirm PMI changes, as TIR

intensity has been shown to correlate reversibly to PMI changes (Viitala et al. 2013).

Unfortunately, the instrumentation used in this study does not provide this data.

11.3.4 Concluding remarks

No straightforward comparison between PMI and PAP sensograms should be done.

When working with living cells, every SPR assay shows a slightly unique response. For

strong SPR responses, such as with PEI/DNA, the results are more consistent and repet-

itive: small variations in SPR signal caused by cellular responses to other factors than

the actual sample are much weaker than the signal caused by PEI/DNA interaction with

cells. For exosome samples causing a weaker response, the signal baseline shifting is

more significant in relation to the observed response. At the very beginning, there is al-

ways drifting of PAP and PMI. Even if this drift settles within 15-30 minutes, constant

increase or decrease in PAP and PMI often exist when samples are injected. For this

Page 77: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

71

reason, meaningful signals are distinguished from the irrelevant ones and only repre-

sentative data is presented. Also, to make the SPR responses easier to interpret, some

data processing has been done to compensate for signal drifting. For example the PAP

sensogram of the platelet exosome sample presented in figure 34 b) shows no change

after the second injection, whereas in the original sensogram the PAP increases slightly

due to baseline drift. Thus, the continuously decreasing PMI of platelet sample after

second injection (Figure 37 b)) actually corresponds reversibly to PAP sensogram. If

these sensograms are compared as such, it seems that PMI changes and PAP does not –

an interpretation that is not correct.

To confirm that these SPR responses are not caused by nonspecific adhesion between

exosomes and cells, or exosomes and gold substrate, experiments with prostate exo-

somes on a sensor with a monolayer of dead cells and on an empty sensor were per-

formed. PAP, TIR angle and PMI sensograms corresponded to the results acquired with

plain medium injections on a sensor with a monolayer of dead cells, or on an empty

sensor (Appendix figures A2, A3 and A4). This confirms that the responses observed

with exosome samples are actually the result of exosomes interacting with the living

cell monolayer. In addition, if PAP increase would be the result of exosomes interacting

with the gold substrate, also the PMI should increase, as the increasing exosome con-

centration on sensor surface is expected to scatter more light.

As mentioned earlier, before injecting the samples, PAP always shows either an in-

crease or a decrease and is not included in the sensograms presented here. When observ-

ing the PMI and TIR angle changes before sample injections, it was evident that the ini-

tial PMI change showed shifting opposite to that of PAP, while the TIR angle shifted to

the same direction as PAP. For control samples with dead cells or with a blank sensor,

the initial PMI, PAP and TIR angle all shifted to the same direction or did not change at

all (Appendix figure A5). Thus, the opposite PMI and PAP responses seem to be char-

acteristic for a living cell layer and possibly specific for LNCaP cells. In addition, the

distinct responses to exosomes of different origin might be specific for this cell type.

This cell line has already shown unique responses toward EGF stimulation when com-

pared to other carcinoma cells (Hiragun et al. 2012). Further experiments with different

Page 78: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

72

cell types and LNCaP cells exposed to exosomes of varying origins are needed to clari-

fy this matter.

The SPR measurements most likely sense very little of the bare gold surface. The wave-

length used in these experiments is 783 nm, resulting in a propagation distance of sur-

face plasmons of approximately 10 µm (Johansen et al. 2000; Golosovsky et al. 2009).

The LNCaP cell has an elongated shape of approximately 100 µm in length and 10-20

µm in width. As the sensors are grown confluent before experimentation, a single sur-

face plasmon resonating and propagating at the gold surface most likely senses only the

cell membrane covered surface. The light beam is approximately 1 mm in width with

several hundreds to thousands of cells colonizing this area. Therefore, SPR sensograms

represent the mean response of several cells. By using the SPR imaging method, sen-

sograms could be resolved within single cell level (Wang et al. 2012a). This would pro-

vide deeper understanding whether responses originate from a single cell, or if i.e. PMI

decrease is a bulk response of the whole cell monolayer. Furthermore, using SPRI, cel-

lular responses from different parts of a single cell could be detected.

The temperature of the medium in our experiments was 37 °C to ensure best possible

environmental conditions for cells during the measurements. As shown with PEI/DNA

interaction on HeLa cells, lowering the temperature enhances the SPR response (Silén

2013). On the other hand, clathrin mediated endocytosis ceases at lower temperatures

(Yashunsky et al. 2009). By using variable temperatures, stronger responses and further

information on exosome internalization mechanisms could be obtained. In addition, in

most studies, a constant flow is used as opposed to the static medium environment in

our experiments. Introducing the sample with continuous flow would ensure no bulk

effect to occur, as can happen when injecting the whole sample at once. However, this

could lead to decreased signal in the SPR response, as exosome concentration interact-

ing with the cells at once is lower. Consequently, purer exosome samples with higher

concentration would be required. Also, before using different temperatures and continu-

ous flow, it must be confirmed that cells can survive under these conditions.

Page 79: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

73

Living cell sensing with SPR is a relatively new field of research, and cellular events

responsible for SPR signals are not yet fully understood. It is, however, widely accepted

that the increase in PAP can be associated to increased amount of mass within evanes-

cent field (Tudos and Schasfoort 2008). The mass increase can be further affirmed by a

simultaneous and parallel shift in TIR angle (Viitala et al. 2013). The decrease in PMI

indicates the formation of more homogenous environment with less light scattering sur-

faces within the evanescent field (Yashunsky et al. 2010b). Despite these facts, the in-

tracellular or cell monolayer reactions responsible for the SPR signals in this study can-

not be unambiguously explained, but more experiments are required to understand the

results in depth.

Exosome sample preparation offers a great challenge. The common protocol for exo-

some purification is based on separation by ultracentrifugation (Bijnsdorp et al. 2013;

Ramteke et al. 2013). With this protocol, the produced samples are not always homoge-

nous in size, can contain other vesicles that are similar in their size and the obtained

quantities are small and variable. However, other purification methods do not necessari-

ly offer considerable improvement and they can be more complicated and laborious

(Tauro et al. 2012). The research on exosomes is still in an early phase of development.

SPR is an interesting option for exosome studies. It is a potential tool for investigating

the different exosome interactions with different cells, as different transportation routes,

endocytic mechanisms and intracellular events produce different responses in SPR.

However, for SPR to become a more feasible method for exosome studies, new tech-

nologies for the production of homogenous exosome samples in large quantities are

needed. With more concentrated exosome samples clearer responses in SPR could be

obtained. In addition, with samples of varying concentration, kinetic information on ex-

osome-cell reaction could be acquired. With exosome samples of better quality, SPR

can be used to investigate exosome-cell interactions in depth.

Page 80: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

74

12 FUTURE PROSPECTS

To gain deeper understanding on cell-exosome interactions, different experimental set-

ups with different detection formats are needed. It is evident that living cells are the ma-

jor source of signal variability in our experiments. An approach where cells are not cul-

tured on the sensor surface would simplify the experimental setup by removing all dis-

crepancies in SPR signals originating from the cells. By using a more pure and simple

running buffer, without contaminants found in the growth medium used in our experi-

ments, it should be possible to determine the degree of exosome enrichment of the sam-

ple liquid with SPR. Thus, culturing LNCaP cells on top of the flow cell chamber and

detecting exosome depletion from the liquid, information on the kinetics of exosome

uptake by the cells could be obtained. Alternatively, detecting the rate of exosome en-

richment of the liquid, determination of exosome secretion of the cells could be made.

Furthermore, as exosomes are enriched with surface proteins for recognition of targeted

cells, with the use of specific antibodies functionalized on sensor surface, these surface

proteins can be exploited to enhance the sensitivity of the assay or to capture specific

exosomes.

13 CONCLUSIONS

LNCaP and PC-3 cells can be cultured on gold-coated sensor slides, and they can with-

stand the stress under SPR measurement. Internalization of PEI/DNA nanoparticles into

cells could be observed with SPR. Also, platelet and prostate exosomes showed re-

sponses corresponding to PEI/DNA signals and thus, these signals are most likely

caused by exosome uptake into the cells. However, the results are difficult to interpret

due to signal baseline drifting and response variability in living cells. The use of SPR in

living cell sensing is still under development, and more work is needed to understand

how different SPR signals relate to cell responses. Use of constant flow, different tem-

perature conditions and more refined sample purification with different sample concen-

trations should provide better understanding of exosome interactions with cells.

Page 81: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

75

REFERENCES

Abdel-Raheem IT, Hide I, Yanase Y, Shigemoto-Mogami Y, Sakai N, Shirai Y et al.: Protein kinase C-α mediates TNF release process in RBL-2H3 mast cells. Br J Pharmacol 145: 415-423, 2005

Akers J, Gonda D, Kim R, Carter B and Chen C: Biogenesis of extracellular vesicles (EV): exosomes, microvesicles, retrovirus-like vesicles, and apoptotic bodies. J Neurooncol 113: 1-11, 2013

Akimoto T, Sasaki S, Ikebukuro K and Karube I: Effect of incident angle of light on sensitivity and detection limit for layers of antibody with surface plasmon resonance spectroscopy. Biosens Bioelectron 15: 355-362, 2000

Auger-Messier M, Arguin G, Chaloux B, Leduc R, Escher E and Guillemette G: Down-Regulation of Inositol 1,4,5-Trisphosphate Receptor in Cells Stably Expressing the Constitutively Active Angiotensin II N111G-AT1 Receptor. Mol Endocrinol 18: 2967-2980, 2004

Baumgarten S and Robelek R: Surface plasmon resonance (SPR) sensors for the rapid, sensitive detection of the cellular response to osmotic stress. Sensor Actuat B-Chem 156: 798-804, 2011

Bijnsdorp I, Geldof A, Lavaei M, Piersma S, van Moorselaar R and Jimenez C: Exosomal ITGA3 interferes with non-cancerous prostate cell functions and is increased in urine exosomes of metastatic prostate cancer patients. J Extracell Vesicles 2: 1-10, 2013

Bobrie A, Colombo M, Raposo G and Théry C: Exosome Secretion: Molecular Mechanisms and Roles in Immune Responses. Traffic 12: 1659-1668, 2011

Bombera R, Leroy L, Livache T and Roupioz Y: DNA-directed capture of primary cells from a complex mixture and controlled orthogonal release monitored by SPR imaging. Biosens Bioelectron 33: 10-16, 2012

Boussif O, Lezoualc'h F, Zanta MA, Mergny MD, Scherman D, Demeneix B et al.: A versatile vector for gene and oligonucleotide transfer into cells in culture and in vivo: polyethylenimine. P Natl Acad Sci USA 92: 7297-7301, 1995

Briske-Anderson MJ, Finley JW and Newman SM: The influence of culture time and passage number on the morphological and physiological development of Caco-2 cells. Proc Soc Exp Biol Med 214: 248-257, 1997

Brockman JM, Frutos AG and Corn RM: A Multistep Chemical Modification Procedure To Create DNA Arrays on Gold Surfaces for the Study of Protein−DNA Interactions with Surface Plasmon Resonance Imaging. J Am Chem Soc 121: 8044-8051, 1999

Page 82: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

76

Burridge K, Fath K, Kelly T, Nuckolls G and Turner C: Focal Adhesions: Transmembrane Junctions Between the Extracellular Matrix and the Cytoskeleton. Annu.Rev.Cell Biol. 4: 487-525, 1988

Caby M, Lankar D, Vincendeau-Scherrer C, Raposo G and Bonnerot C: Exosomal-like vesicles are present in human blood plasma. Int Immunol 17: 879-887, 2005

Canadian Cancer Society’s Steering Committee on Cancer Statistics: Canadian Cancer Statistics 2012. Canadian Cancer Society, Toronto, ON 2012

Castillo AM, Lagunes R, Urbán M, Frixione E and Meza I: Myosin II-actin interaction in MDCK cells: role in cell shape changes in response to Ca2+ variations. J Muscle Res Cell Motil 19: 557-74, 1998

Chabot V, Cuerrier CM, Escher E, Aimez V, Grandbois M and Charette PG: Biosensing based on surface plasmon resonance and living cells. Biosens Bioelectron 24: 1667-1673, 2009

Chabot V, Miron Y, Charette PG and Grandbois M: Identification of the molecular mechanisms in cellular processes that elicit a surface plasmon resonance (SPR) response using simultaneous surface plasmon-enhanced fluorescence (SPEF) microscopy. Biosens Bioelectron 50: 125-131, 2013

Chachisvilis M, Zhang Y and Frangos JA: G protein-coupled receptors sense fluid shear stress in endothelial cells. P Natl Acad Sci USA 103: 15463-15468, 2006

Chen K, Li Y, Kim M, Li S, Yuan S, Chien S et al.: Mechanotransduction in Response to Shear Stress: Roles of Receptor Tyrosine Kinases, Integrins, and Shc. J Biol Chem 274: 18393-18400, 1999

Chen WT and Singer SJ: Immunoelectron microscopic studies of the sites of cell-substratum and cell-cell contacts in cultured fibroblasts. J Cell Biol 95: 205-222, 1982

Chiang H-, Wang Y-, Leung PT and Tse WS: A theoretical model for the temperature-dependent sensitivity of the optical sensor based on surface plasmon resonance. Opt Commun 188: 283-289, 2001

Choi J, Park K, Lee D, Lee W and Lee WH: Cell immobilization using self-assembled synthetic oligopeptide and its application to biological toxicity detection using surface plasmon resonance. Biosens Bioelectron 20: 2300-2305, 2005

Cocucci E, Racchetti G and Meldolesi J: Shedding microvesicles: artefacts no more. Trends Cell Biol 19: 43-51, 2009

Cuerrier CM, Chabot V, Vigneux S, Aimez V, Escher E, Gobeil F et al.: Surface Plasmon Resonance Monitoring of Cell Monolayer Integrity: Implication of Signaling Pathways Involved in Actin-Driven Morphological Remodeling. Cell Mol Bioeng 1: 229-239, 2008

Page 83: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

77

Cuerrier CM, Benoit M, Guillemette G, Jr Gobeil F and Grandbois M: Real-time monitoring of angiotensin II-induced contractile response and cytoskeleton remodeling in individual cells by atomic force microscopy. Pfluc Arch Eur J Phy 457: 1361-72, 2009

Douglas RM and Haddad GG: Invited Review: Effect of oxygen deprivation on cell cycle activity: a profile of delay and arrest. J Appl Phys 94: 2068-2083, 2003

Ehrlich M, Boll W, van Oijen A, Hariharan R, Chandran K, Nibert ML et al.: Endocytosis by Random Initiation and Stabilization of Clathrin-Coated Pits. Cell 118: 591-605, 2004

Esquenet M, Swinnen JV, Heyns W and Verhoeven G: LNCaP prostatic adenocarcinoma cells derived from low and high passage numbers display divergent responses not only to androgens but also to retinoids. J Steroid Biochem Mol Biol 62: 391-399, 1997

Fang Y, Ferrie AM, Fontaine NH, Mauro J and Balakrishnan J: Resonant Waveguide Grating Biosensor for Living Cell Sensing. Biophys J 91: 1925-1940, 2006

Ferlay J, Steliarova-Foucher E, Lortet-Tieulent J, Rosso S, Coebergh JWW, Comber H et al.: Cancer incidence and mortality patterns in Europe: Estimates for 40 countries in 2012. Eur J Cancer 49: 1374-1403, 2013

Fratamico PM, Strobaugh TP, Medina MB and Gehring AG: Detection of Escherichia coli 0157:H7 using a surface plasmon resonance biosensor. Biotechnol Tech 12: 571-576, 1998

Giebel KF, Bechinger C, Herminghaus S, Riedel M, Leiderer P, Weiland U et al.: Imaging of cell/substrate contacts of living cells with surface plasmon resonance microscopy. Biophys J 76: 509-516, 1999

Godbey WT, Wu KK and Mikos AG: Tracking the intracellular path of poly(ethylenimine)/DNA complexes for gene delivery. P Natl Acad Sci USA 96: 5177-5181, 1999

Golosovsky M, Lirtsman V, Yashunsky V, Davidov D and Aroeti B: Midinfrared surface-plasmon resonance: A novel biophysical tool for studying living cells. J Appl Phys 105: 102036, 2009

González JE and Negulescu PA: Intracellular detection assays for high-throughput screening. Curr Opin Biotechnol 9: 624-631, 1998

Gordon JG and Ernst S: Surface plasmons as a probe of the electrochemical interface. Surf Sci 101: 499-506, 1980

Hanzlíková M, Ruponen M, Galli E, Raasmaja A, Aseyev V, Tenhu H et al.: Mechanisms of polyethylenimine-mediated DNA delivery: free carrier helps to overcome the barrier of cell-surface glycosaminoglycans. J Gene Med 13: 402-409, 2011

Page 84: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

78

Hayes J and Barry M: Screening for Prostate Cancer With the Prostate-Specific Antigen Test: A Review of Current Evidence. JAMA 311: 1143-1149, 2014

He R, Chang G, Wu H, Lin C, Chiu K, Su Y et al.: Enhanced live cell membrane imaging using surface plasmon-enhanced total internal reflection fluorescence microscopy. Opt Express 14: 9307-9316, 2006

Hide M, Tsutsui T, Sato H, Nishimura T, Morimoto K, Yamamoto S et al.: Real-Time Analysis of Ligand-Induced Cell Surface and Intracellular Reactions of Living Mast Cells Using a Surface Plasmon Resonance-Based Biosensor. Anal Biochem 302: 28-37, 2002

Hiragun T, Yanase Y, Kose K, Kawaguchi T, Uchida K, Tanaka S et al.: Surface plasmon resonance-biosensor detects the diversity of responses against epidermal growth factor in various carcinoma cell lines. Biosens Bioelectron 32: 202-207, 2012

Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT and De Bruijn EA: Vascular Endothelial Growth Factor and Angiogenesis. Pharmacol Rev 56: 549-580, 2004

Homola J: Surface Plasmon Resonance Based Sensors. Springer Series on Chemical Sensors and Biosensors. Vol. 4. Series Edit. Wolfbeis OS. Springer, Berlin 2006

Hopkins AL and Groom CR: Opinion: The druggable genome. Nat Rev Drug Discov 1: 727-730, 2002

Horii M, Shinohara H, Iribe Y and Suzuki M: Living cell-based allergen sensing using a high resolution two-dimensional surface plasmon resonance imager. Analyst 136: 2706-2711, 2011

Illinger D, Poindron P and Kuhry J: Fluid phase endocytosis investigated by fluorescence with trimethylamino-diphenylhexatriene in L929 cells; the influence of temperature and of cytoskeleton depolymerizing drugs. Biol Cell 73: 131-138, 1991

Iribe Y, Shinohara H and Suzuki M: Label-free detection of B and T cell responses by using high resolution 2D-SPR imaging sensor. The 14th International Conference on Miniaturized Systems for Chemistry and Life Sciences 1646-1648, 2010

Ivic L, Zhang C, Zhang X, Yoon SO and Firestein S: Intracellular trafficking of a tagged and functional mammalian olfactory receptor. J Neurobiol 50: 56-68, 2002

Izzard CS and Lochner LR: Cell-to-substrate contacts in living fibroblasts: an interference reflexion study with an evaluation of the technique. J Cell Sci 21: 129-159, 1976

Jansen FH, Krijgsveld J, van Rijswijk A, van den Bemd G, van den Berg MS, van Weerden WM et al.: Exosomal Secretion of Cytoplasmic Prostate Cancer Xenograft-derived Proteins. Mol Cell Proteomics 8: 1192-1205, 2009

Page 85: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

79

Johansen K, Arwin H, Lundström I and Liedberg B: Imaging surface plasmon resonance sensor based on multiple wavelengths: Sensitivity considerations. Rev Sci Instrum 71: 3530-3538, 2000

Johnson AE: Fluorescence Approaches for Determining Protein Conformations, Interactions and Mechanisms at Membranes. Traffic 6: 1078-1092, 2005

Jung T, Castellana D, Klingbeil P, Cuesta Hernández I, Vitacolonna M, Orlicky D et al.: CD44v6 dependence of premetastatic niche preparation by exosomes. Neoplasia 11: 1093-1105, 2009

Kooyman RPH: Physics of Surface Plasmon Resonance. Handbook of Surface Plasmon Resonance. p. 15-34, 1st edition. Edit. Schasfoort RBM and Tudos AJ. The Royal Society of Chemistry, Cambridge 2008

Kosaihira A and Ona T: Rapid and quantitative method for evaluating the personal therapeutic potential of cancer drugs. Anal Bioanal Chem 391: 1889-1897, 2008

Kretschmann E and Raether H: Radiative decay of non radiative surface plasmons excited by light (Surface plasma waves excitation by light and decay into photons applied to nonradiative modes). Z Naturforsch A 23: 2135, 1968

Lasser C, Seyed Alikhani V, Ekstrom K, Eldh M, Torregrosa Paredes P, Bossios A et al.: Human saliva, plasma and breast milk exosomes contain RNA: uptake by macrophages. J Transl Med 9: 1-8, 2011

Lee E, Yoo G, Jose J, Kang M, Song S and Pyun J: SPR biosensor based on immobilized E.coli cells with autodisplayed Z-domains. Biochip J 6: 221-228, 2012

Lee JY, Ko HJ, Lee SH and Park TH: Cell-based measurement of odorant molecules using surface plasmon resonance. Enzyme Microb Technol 39: 375-380, 2006

Li Y, Lee HJ and Corn RM: Fabrication and characterization of RNA aptamer microarrays for the study of protein–aptamer interactions with SPR imaging. Nucleic Acids Res 34: 6416-6424, 2006

Lin H, Hu Y, Yang L, Altuwaijri S, Chen Y, Kang H et al.: Suppression Versus Induction of Androgen Receptor Functions by the Phosphatidylinositol 3-Kinase/Akt Pathway in Prostate Cancer LNCaP Cells with Different Passage Numbers. J Biol Chem 278: 50902-50907, 2003

Lirtsman V, Golosovsky M and Davidov D: Infrared surface plasmon resonance technique for biological studies. J Appl Phys 103: 0147021-0147025, 2008

Liu C, Lei T, Ino K, Matsue T, Tao N and Li C: Real-time monitoring biomarker expression of carcinoma cells by surface plasmon resonance biosensors. Chem Commun 48: 10389-10391, 2012

Liu T, Mendes D and Berkman C: Functional prostate-specific membrane antigen is enriched in exosomes from prostate cancer cells. Int J Oncol 44: 918-922, 2014

Page 86: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

80

Llorente A, van Deurs B and Sandvig K: Cholesterol regulates prostasome release from secretory lysosomes in PC-3 human prostate cancer cells. Eur J Cell Biol 86: 405-415, 2007

Lu Q, Zhang J, Allison R, Gay H, Yang W, Bhowmick NA et al.: Identification of extracellular δ-catenin accumulation for prostate cancer detection. Prostate 69: 411-418, 2009

Maier SA: Plasmonics: Fundamentals and Applications. 1st edition. Springer, New York 2007

Mäkinen T, Auvinen A, Hakama M, Stenman U and Tammela TLJ: Acceptability and complications of prostate biopsy in population-based PSA screening versus routine clinical practice: a prospective, controlled study. Urology 60: 846-850, 2002

Maltais J, Denault J, Gendron L and Grandbois M: Label-free monitoring of apoptosis by surface plasmon resonance detection of morphological changes. Apoptosis 17: 916-925, 2012

Mannelli I, Courtois V, Lecaruyer P, Roger G, Millot MC, Goossens M et al.: Surface plasmon resonance imaging (SPRI) system and real-time monitoring of DNA biochip for human genetic mutation diagnosis of DNA amplified samples. Sensors Actuators B: Chem 119: 583-591, 2006

Martins VR, Dias MS and Hainaut P: Tumor-cell-derived microvesicles as carriers of molecular information in cancer. Curr Opin Oncol 25: 66-75, 2013

Mauriz E, Carbajo-Pescador S, Ordonez R, Garcia-Fernandez M, Mauriz JL, Lechuga LM et al.: On-line surface plasmon resonance biosensing of vascular endothelial growth factor signaling in intact-human hepatoma cell lines. Analyst 139: 1426-1435, 2014

Michaelis S, Wegener J and Robelek R: Label-free monitoring of cell-based assays: Combining impedance analysis with SPR for multiparametric cell profiling. Biosens Bioelectron 49: 63-70, 2013

Milgram S, Cortes S, Villiers M, Marche P, Buhot A, Livache T et al.: On chip real time monitoring of B-cells hybridoma secretion of immunoglobulin. Biosens Bioelectron 26: 2728-2732, 2011

Mir T and Shinohara H: Real-time monitoring of cell response to drug stimulation by 2D-SPR Sensor: An approach to study neuronal differentiation. 14th International Meeting on Chemical Sensors 863-865, 2012

Mizuguchi T, Uchimura H, Kataoka H, Akaji K, Kiso Y and Saito K: Intact-cell-based surface plasmon resonance measurements for ligand affinity evaluation of a membrane receptor. Anal Biochem 420: 185-187, 2012

Nelson BP, Grimsrud TE, Liles MR, Goodman RM and Corn RM: Surface Plasmon Resonance Imaging Measurements of DNA and RNA Hybridization Adsorption onto DNA Microarrays. Anal Chem 73: 1-7, 2001

Page 87: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

81

Nelson BP, Liles MR, Frederick KB, Corn RM and Goodman RM: Label-free detection of 16S ribosomal RNA hybridization on reusable DNA arrays using surface plasmon resonance imaging. Environ Microbiol 4: 735-743, 2002

Nishijima H, Kosaihira A, Shibata J and Ona T: Development of Signaling Echo Method for Cell-based Quantitative Efficacy Evaluation of Anti-cancer Drugs in Apoptosis without Drug Presence Using High-precision Surface Plasmon Resonance Sensing. Anal Sci 26: 529-534, 2010

Ono M and Kuwano M: Molecular Mechanisms of Epidermal Growth Factor Receptor (EGFR) Activation and Response to Gefitinib and Other EGFR-Targeting Drugs. Clin Cancer Res 12: 7242-7251, 2006

Otto A: Excitation of nonradiative surface plasma waves in silver by the method of frustrated total reflection. Z Phys 216: 398-410, 1968

Peinado H, Aleckovic M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G et al.: Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med 18: 883-891, 2012

Poliakov A, Spilman M, Dokland T, Amling CL and Mobley JA: Structural heterogeneity and protein composition of exosome-like vesicles (prostasomes) in human semen. Prostate 69: 159-167, 2009

Qian ZM, Li H, Sun H and Ho K: Targeted Drug Delivery via the Transferrin Receptor-Mediated Endocytosis Pathway. Pharmacol Rev 54: 561-587, 2002

Qu J, Qu X, Zhao M, Teng Y, Zhang Y, Hou K et al.: Gastric cancer exosomes promote tumour cell proliferation through PI3K/Akt and MAPK/ERK activation. Dig Liver Dis 41: 875-880, 2009

Quinn JG, O'Neill S, Doyle A, McAtamney C, Diamond D, MacCraith BD et al.: Development and Application of Surface Plasmon Resonance-Based Biosensors for the Detection of Cell–Ligand Interactions. Anal Biochem 281: 135-143, 2000

Raether H: Surface Plasmons on Smooth and Rough Surfaces and on Gratings. 1st edition. Springer, Berlin 1986

Raj DAA, Fiume I, Capasso G and Pocsfalvi G: A multiplex quantitative proteomics strategy for protein biomarker studies in urinary exosomes. Kidney Int 81: 1263-1272, 2012

Ramteke A, Ting H, Agarwal C, Mateen S, Somasagara R, Hussain A et al.: Exosomes secreted under hypoxia enhance invasiveness and stemness of prostate cancer cells by targeting adherens junction molecules. Mol Carcinog 1-12, 2013

Reinsch S and Karsenti E: Orientation of spindle axis and distribution of plasma membrane proteins during cell division in polarized MDCKII cells. J Cell Biol 126: 1509-1526, 1994

Page 88: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

82

Rémy-Kristensen A, Clamme J, Vuilleumier C, Kuhry J and Mély Y: Role of endocytosis in the transfection of L929 fibroblasts by polyethylenimine/DNA complexes. Biochim Biophys Acta 1514: 21-32, 2001

Robelek R and Wegener J: Label-free and time-resolved measurements of cell volume changes by surface plasmon resonance (SPR) spectroscopy. Biosens Bioelectron 25: 1221-1224, 2010

Rothenhäusler B and Knoll W: Surface–-plasmon microscopy. Nature 332: 615-617, 1988

Samaj J, Baluska F, Voigt B, Schlicht M and Menzel D: Endocytosis, actin cytoskeleton, and signaling. Plant Physiol 135: 1150-1161, 2004

Schasfoort RBM and McWhirter A: SPR Instrumentation. Handbook of Surface Plasmon Resonance. p. 35-80, 1st edition. Edit. Schasfoort RBM and Tudos AJ. The Royal Society of Chemistry, Cambridge 2008

Schwartz GK and Shah MA: Targeting the cell cycle: A new approach to cancer therapy. J Clin Oncol 23: 9408-9421, 2005

Shifrin Y, Kirschner J, Geiger B and Rosenshine I: Enteropathogenic Escherichia coli induces modification of the focal adhesions of infected host cells. Cell Microbiol 4: 235-243, 2002

Siegel R, Naishadham D and Jemal A: Cancer statistics, 2013. CA Cancer J Clin 63: 11-30, 2013

Silén T: Dissertation. In vitro screening of new ternary polymeric nanoparticles for gene delivery with cell surfaces. University of Helsinki, Faculty of Pharmacy, Division of Biopharmaceutics and Pharmacokinetics, 2013

Siraganian RP: Mast cell signal transduction from the high-affinity IgE receptor. Curr Opin Immunol 15: 639-646, 2003

Smith EA, Thomas WD, Kiessling LL and Corn RM: Surface Plasmon Resonance Imaging Studies of Protein-Carbohydrate Interactions. J Am Chem Soc 125: 6140-6148, 2003

Sokolova V, Ludwig A, Hornung S, Rotan O, Horn PA, Epple M et al.: Characterisation of exosomes derived from human cells by nanoparticle tracking analysis and scanning electron microscopy. Colloid Surface B 87: 146-150, 2011

Stahelin RV: Surface plasmon resonance: a useful technique for cell biologists to characterize biomolecular interactions. Mol Biol Cell 24: 883-886, 2013

Steltenkamp S, Rommel C, Wegener J and Janshoff A: Membrane stiffness of animal cells challenged by osmotic stress. Small 2: 1016-1020, 2006

Page 89: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

83

Steyer JA and Almers W: A Real-Time View of Life within 100 Nm of the Plasma Membrane. Nat Rev Mol Cell Bio 2: 268-275, 2001

Suraniti E, Sollier E, Calemczuk R, Livache T, Marche PN, Villiers M et al.: Real-time detection of lymphocytes binding on an antibody chip using SPR imaging. Lab Chip 7: 1206-1208, 2007

Suzuki H, Yanase Y, Tsutsui T, Ishii K, Hiragun T and Hide M: Applying Surface Plasmon Resonance to Monitor the IgE-Mediated Activation of Human Basophils. Allergol Int 57: 347-358, 2008

Tanaka M, Hiragun T, Tsutsui T, Yanase Y, Suzuki H and Hide M: Surface plasmon resonance biosensor detects the downstream events of active PKCβ in antigen-stimulated mast cells. Biosens Bioelectron 23: 1652-1658, 2008

Tauro BJ, Greening DW, Mathias RA, Ji H, Mathivanan S, Scott AM et al.: Comparison of ultracentrifugation, density gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell line LIM1863-derived exosomes. Methods 56: 293-304, 2012

Tawil N, Sacher E, Mandeville R and Meunier M: Surface plasmon resonance detection of E. coli and methicillin-resistant S. aureus using bacteriophages. Biosens Bioelectron 37: 24-29, 2012

Thakur BK, Zhang H, Becker A, Matei I, Huang Y, Costa-Silva B et al.: Double-stranded DNA in exosomes: a novel biomarker in cancer detection. Cell Res 2014

Tudos AJ and Schasfoort RBM: Introduction to Surface Plasmon Resonance. Handbook of Surface Plasmon Resonance. p. 1-14, 1st edition. Edit. Schasfoort RBM and Tudos AJ. The Royal Society of Chemistry, Cambridge 2008

Vala M, Robelek R, Bocková M, Wegener J and Homola J: Real-time label-free monitoring of the cellular response to osmotic stress using conventional and long-range surface plasmons. Biosens Bioelectron 40: 417-421, 2013

Valadi H, Ekström K, Bossios A, Sjöstrand M, Lee JJ and Lötvall J,O.: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol 9: 654-659, 2007

Viitala T, Granqvist N, Hallila S, Raviña M and Yliperttula M: Elucidating the Signal Responses of Multi-Parametric Surface Plasmon Resonance Living Cell Sensing: A Comparison between Optical Modeling and Drug–MDCKII Cell Interaction Measurements. PLOS ONE 8: 1-13, 2013

Vlassov AV, Magdaleno S, Setterquist R and Conrad R: Exosomes: Current knowledge of their composition, biological functions, and diagnostic and therapeutic potentials. BBA-Gen Subjects 1820: 940-948, 2012

Page 90: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

84

Wahlgren J, Karlson TDL, Brisslert M, Vaziri Sani F, Telemo E, Sunnerhagen P et al.: Plasma exosomes can deliver exogenous short interfering RNA to monocytes and lymphocytes. Nucl Acids Res 40: e130-e130, 2012

Wakao M, Saito A, Ohishi K, Kishimoto Y, Nishimura T, Sobel M et al.: Sugar Chips immobilized with synthetic sulfated disaccharides of heparin/heparan sulfate partial structure. Bioorg Med Chem Lett 18: 2499-2504, 2008

Wang W, Yang Y, Wang S, Nagaraj VJ, Liu Q, Wu J et al.: Label-free measuring and mapping of binding kinetics of membrane proteins in single living cells. Nature Chem 4: 846-853, 2012a

Wang W, Wang S, Liu Q, Wu J and Tao N: Mapping Single-Cell–Substrate Interactions by Surface Plasmon Resonance Microscopy. Langmuir 28: 13373-13379, 2012b

Wang Y, Shyy JY and Chien S: Fluorescence Proteins, Live-Cell Imaging, and Mechanobiology: Seeing Is Believing. Annu Rev Biomed Eng 10: 1-38, 2008

Waswa JW, Debroy C and Irudayaraj J: Rapid Detection of Salmonella enteritidis and Escherichia coli Using Surface Plasmon Resonance Biosensor. J Food Process Eng 29: 373-385, 2006

Waswa J, Irudayaraj J and DebRoy C: Direct detection of E. Coli O157:H7 in selected food systems by a surface plasmon resonance biosensor. LWT-Food Sci Technol 40: 187-192, 2007

Watts HJ, Lowe CR and Pollard-Knight D: Optical Biosensor for Monitoring Microbial Cells. Anal Chem 66: 2465-2470, 1994

Wegner GJ, Lee HJ, Marriott G and Corn RM: Fabrication of Histidine-Tagged Fusion Protein Arrays for Surface Plasmon Resonance Imaging Studies of Protein−Protein and Protein−DNA Interactions. Anal Chem 75: 4740-4746, 2003

Witwer K, Buzás E, Bemis L, Bora A, Lässer C, Lötvall J et al.: Standardization of sample collection, isolation and analysis methods in extracellular vesicle research. J Extracell Vesicles 2: 1-25, 2013

Wong CL, Ho HP, Suen YK, Kong SK, Chen QL, Yuan W et al.: Real-time protein biosensor arrays based on surface plasmon resonance differential phase imaging. Biosens Bioelectron 24: 606-612, 2008

Yanase Y, Hiragun T, Yanase T, Kawaguchi T, Ishii K and Hide M: Application of SPR Imaging Sensor for Detection of Individual Living Cell Reactions and Clinical Diagnosis of Type I Allergy. Allergol Int 62: 163-169, 2013

Yanase Y, Suzuki H, Tsutsui T, Uechi I, Hiragun T, Mihara S et al.: Living cell positioning on the surface of gold film for SPR analysis. Biosens Bioelectron 23: 562-567, 2007a

Page 91: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

85

Yanase Y, Suzuki H, Tsutsui T, Hiragun T, Kameyoshi Y and Hide M: The SPR signal in living cells reflects changes other than the area of adhesion and the formation of cell constructions. Biosens Bioelectron 22: 1081-1086, 2007b

Yanase Y, Hiragun T, Kaneko S, Gould HJ, Greaves MW and Hide M: Detection of refractive index changes in individual living cells by means of surface plasmon resonance imaging. Biosens Bioelectron 26: 674-681, 2010

Yanase Y, Hiragun T, Yanase T, Kawaguchi T, Ishii K and Hide M: Evaluation of peripheral blood basophil activation by means of surface plasmon resonance imaging. Biosens Bioelectron 32: 62-68, 2012

Yashunsky V, Lirtsman V, Golosovsky M, Davidov D and Aroeti B: Real-time monitoring of epithelial cell-cell and cell-substrate interactions by infrared surface plasmon spectroscopy. Biophys J 99: 4028-4036, 2010b

Yashunsky V, Shimron S, Lirtsman V, Weiss AM, Melamed-Book N, Golosovsky M et al.: Real-Time Monitoring of Transferrin-Induced Endocytic Vesicle Formation by Mid-Infrared Surface Plasmon Resonance. Biophys J 97: 1003-1012, 2009

Yashunsky V, Marciano T, Lirtsman V, Golosovsky M, Davidov D and Aroeti B: Real-Time Sensing of Cell Morphology by Infrared Waveguide Spectroscopy. PLOS ONE 7: 1-10, 2012a

Yashunsky V, Lirtsman V, Zilbershtein A, Bein A, Schwartz B, Aroeti B et al.: Surface plasmon-based infrared spectroscopy for cell biosensing. J Biomed Opt 17: 0814091-0814098, 2012b

Yashunsky V, Kharilker L, Zlotkin-Rivkin E, Rund D, Melamed-Book N, Zahavi EE et al.: Real-Time Sensing of EnteropathogenicE. coli-Induced Effects on Epithelial Host Cell Height, Cell-Substrate Interactions, and Endocytic Processes by Infrared Surface Plasmon Spectroscopy. PLOS ONE 8: 1-13, 2013

Yuk JS, Kim H, Jung J, Jung S, Lee S, Kim WJ et al.: Analysis of protein interactions on protein arrays by a novel spectral surface plasmon resonance imaging. Biosens Bioelectron 21: 1521-1528, 2006

Zhang H, Yang L, Zhou B, Wang X, Liu G, Liu W et al.: Investigation of biological cell–protein interactions using SPR sensor through laser scanning confocal imaging–surface plasmon resonance system. Spectrochim Acta A 121: 381-386, 2014

Ziblat R, Lirtsman V, Davidov D and Aroeti B: Infrared surface plasmon resonance: a novel tool for real time sensing of variations in living cells. Biophys J 90: 2592-2599, 2006

Page 92: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

APPENDIX 1 Medium and PEI/DNA sensograms for PC-3 cells.

Figure A1. PAP changes with PC-3 cells for plain medium sample a) and PEI/DNA sample b). Solid arrow indicates injection of sample and dashed arrow marks injection of medium. y-axis shows change of PAP in degrees and x-axis time in minutes.

APPENDIX 2 Control sensograms for PAP, PMI and TIR angle.

Figure A2. PAP sensograms for platelet exosomes and medium sample on pure gold sensor a) and sensor with dead cells b). y-axis shows change of PAP in degrees and x-axis time in minutes.

Page 93: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

Figure A3. TIR angle sensograms for platelet exosomes and medium sample on pure gold sensor a) and sensor with dead cells b). y-axis shows relative change of TIR angle and x-axis time in minutes.

Figure A4. PMI sensograms for platelet exosomes and medium sample on pure gold sensor a) and sensor with dead cells b). y-axis shows the change in reflected light inten-sity and x-axis time in minutes.

Page 94: SURFACE PLASMON RESONANCE IN LIVING CELL SENSING

APPENDIX 3 Initial signal shifting sensograms.

Figure A5. Initial shifting of PAP, PMI and TIR angle on sensor without cells a), sensor with dead cell monolayer b) and sensor with living cell monolayer c). Left y-axis shows change in PAP in degrees, right y-axis is relative TIR angle and PMI change and x axis is time in minutes.