Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic...

8
COMMENTARY Correction for ‘‘Icy insights from emperor penguins,’’ by Colleen Cassady St. Clair and Mark S. Boyce, which appeared in issue 6, February 10, 2009, of Proc Natl Acad Sci USA (106:1691–1692; first published February 4, 2009; 10.1073/pnas.0812940106). The authors note that due to a printer’s error in the Fig. 1 legend, the two panels were credited incorrectly. The corrected legend and its figure appear below. www.pnas.orgcgidoi10.1073pnas.0901386106 Fig. 1. Life cycle for emperor penguins at Terre Ade ´ lie, Antarctica. Survival probability is S i for stage i. Minimum age at first breeding was 3 years; once recruited, birds reproduce annually with probability S a P b , where P b is the proportion of breeders. BS is overall breeding success. Translation of this life cycle into a population projection matrix was described by Jenouvrier et al. (13). In the current article, Jenouvrier et al. (2) link past demographic responses to warm years to IPCC models of projected climate change to predict a persistent decline in the emperor penguin population at Terre Ade ´ lie and a 36% likelihood of extinction by 2100. [Photo of emperor penguins by Gordon Court. Population model reproduced with permission from Jenouvrier et al. (13) (Copyright 2005, Ecological Society of America).] CELL BIOLOGY Correction for ‘‘Reprogramming of murine and human somatic cells using a single polycistronic vector,’’ by Bryce W. Carey, Styliani Markoulaki, Jacob Hanna, Kris Saha, Qing Gao, Maisam Mitalipova, and Rudolf Jaenisch, which appeared in issue 1, January 6, 2009, of Proc Natl Acad Sci USA (106:157–162; first published December 24, 2008; 10.1073pnas.0811426106). The authors note that on page 157, in the right column, the heading, ‘‘Generation of Mouse Ips Cells Using a Single Poly- cistronic Virus,’’ should instead read: ‘‘Generation of Mouse iPS Cells Using a Single Polycistronic Virus.’’ On page 158, right column, in line 5 of the first paragraph and line 6 of the second paragraph, ?8 should instead appear as 8. On page 159, left column, line 9 of the second paragraph, the sentence ‘‘This is two orders of magnitude lower than that of ‘secondary’ fibroblasts or B cells carrying preselected DOX-inducible proviruses (19),’’ should read: ‘‘This is one to two orders of magnitude lower than that of ‘primary’ infected fibroblasts (3, 7).’’ Also on page 159, in the right column, the heading, ‘‘Generation of Human Ips Cells Using a Single Polycistronic Virus,’’ should instead read: ‘‘Generation of Human iPS Cells Using a Single Polycistronic Virus.’’ Finally, on page 160, in the first paragraph of Materials and Methods, the sequence for the P2A peptide “GCCACGAAGCAAGCAG- GAGATGTTGAAGAAAACCCCGG GCCT” should instead read “GCCACGAACTTCTCTCTGTTAAAGCAAGCAGG- AGATGTTGAAGAAAACCCCGGGCCT.” www.pnas.orgcgidoi10.1073pnas.0900269106 CELL BIOLOGY Correction for ‘‘PGC-1 is coupled to HIF-1-dependent gene expression by increasing mitochondrial oxygen consumption in skeletal muscle cells,’’ by Kathleen A. O’Hagan, Sinead Coc- chiglia, Alexander V. Zhdanov, Murtaza M. Tambawala, Eoin P. Cummins, Mona Monfared, Terence A. Agbor, John F. Garvey, Dmitri B. Papkovsky, Cormac T. Taylor, and Bernard B. Allan, which appeared in issue 7, February 17, 2009, of Proc Natl Acad Sci USA (106:2188–2193; first published January 28, 2009; 10.1073pnas.0808801106). The authors note that the author name Murtaza M. Tam- bawala should have appeared as Murtaza M. Tambuwala. The online version has been corrected. The corrected author line appears below. Kathleen A. O’Hagan, Sinead Cocchiglia, Alexander V. Zhdanov, Murtaza M. Tambuwala, Eoin P. Cummins, Mona Monfared, Terence A. Agbor, John F. Garvey, Dmitri B. Papkovsky, Cormac T. Taylor, and Bernard B. Allan www.pnas.orgcgidoi10.1073pnas.0901252106 PNAS March 31, 2009 vol. 106 no. 13 5449 CORRECTIONS Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021 Downloaded by guest on April 28, 2021

Transcript of Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic...

Page 1: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

COMMENTARYCorrection for ‘‘Icy insights from emperor penguins,’’ by ColleenCassady St. Clair and Mark S. Boyce, which appeared in issue 6,February 10, 2009, of Proc Natl Acad Sci USA (106:1691–1692;first published February 4, 2009; 10.1073/pnas.0812940106).

The authors note that due to a printer’s error in the Fig. 1legend, the two panels were credited incorrectly. The correctedlegend and its figure appear below.

www.pnas.org�cgi�doi�10.1073�pnas.0901386106

Fig. 1. Life cycle for emperor penguins at TerreAdelie, Antarctica. Survival probability is Si for stagei. Minimum age at first breeding was 3 years; oncerecruited, birds reproduce annually with probabilitySa Pb, where Pb is the proportion of breeders. BS isoverall breeding success. Translation of this life cycleinto a population projection matrix was described byJenouvrieretal. (13). In thecurrentarticle, Jenouvrieret al. (2) link past demographic responses to warmyears to IPCC models of projected climate change topredict a persistent decline in the emperor penguinpopulation at Terre Adelie and a 36% likelihood ofextinction by 2100. [Photo of emperor penguins byGordon Court. Population model reproduced withpermission from Jenouvrier et al. (13) (Copyright2005, Ecological Society of America).]

CELL BIOLOGYCorrection for ‘‘Reprogramming of murine and human somaticcells using a single polycistronic vector,’’ by Bryce W. Carey,Styliani Markoulaki, Jacob Hanna, Kris Saha, Qing Gao,Maisam Mitalipova, and Rudolf Jaenisch, which appeared inissue 1, January 6, 2009, of Proc Natl Acad Sci USA (106:157–162;first published December 24, 2008; 10.1073�pnas.0811426106).

The authors note that on page 157, in the right column, theheading, ‘‘Generation of Mouse Ips Cells Using a Single Poly-cistronic Virus,’’ should instead read: ‘‘Generation of Mouse iPSCells Using a Single Polycistronic Virus.’’ On page 158, rightcolumn, in line 5 of the first paragraph and line 6 of the secondparagraph, ?8 should instead appear as �8. On page 159, leftcolumn, line 9 of the second paragraph, the sentence ‘‘This is twoorders of magnitude lower than that of ‘secondary’ fibroblasts orB cells carrying preselected DOX-inducible proviruses (19),’’should read: ‘‘This is one to two orders of magnitude lower than thatof ‘primary’ infected fibroblasts (3, 7).’’ Also on page 159, in theright column, the heading, ‘‘Generation of Human Ips Cells Usinga Single Polycistronic Virus,’’ should instead read: ‘‘Generation ofHuman iPS Cells Using a Single Polycistronic Virus.’’ Finally, onpage 160, in the first paragraph of Materials and Methods, thesequence for the P2A peptide “GCCACGAAGCAAGCAG-GAGATGTTGAAGAAAACCCCGG GCCT” should insteadread “GCCACGAACTTCTCTCTGTTAAAGCAAGCAGG-AGATGTTGAAGAAAACCCCGGGCCT.”

www.pnas.org�cgi�doi�10.1073�pnas.0900269106

CELL BIOLOGYCorrection for ‘‘PGC-1� is coupled to HIF-1�-dependent geneexpression by increasing mitochondrial oxygen consumption inskeletal muscle cells,’’ by Kathleen A. O’Hagan, Sinead Coc-chiglia, Alexander V. Zhdanov, Murtaza M. Tambawala, Eoin P.Cummins, Mona Monfared, Terence A. Agbor, John F. Garvey,Dmitri B. Papkovsky, Cormac T. Taylor, and Bernard B. Allan,which appeared in issue 7, February 17, 2009, of Proc Natl AcadSci USA (106:2188–2193; first published January 28, 2009;10.1073�pnas.0808801106).

The authors note that the author name Murtaza M. Tam-bawala should have appeared as Murtaza M. Tambuwala. Theonline version has been corrected. The corrected author lineappears below.

Kathleen A. O’Hagan, Sinead Cocchiglia, Alexander V.Zhdanov, Murtaza M. Tambuwala, Eoin P. Cummins,Mona Monfared, Terence A. Agbor, John F. Garvey,Dmitri B. Papkovsky, Cormac T. Taylor, and Bernard B.Allan

www.pnas.org�cgi�doi�10.1073�pnas.0901252106

PNAS � March 31, 2009 � vol. 106 � no. 13 � 5449

CORR

ECTI

ON

S

Dow

nloa

ded

by g

uest

on

Apr

il 28

, 202

1 D

ownl

oade

d by

gue

st o

n A

pril

28, 2

021

Dow

nloa

ded

by g

uest

on

Apr

il 28

, 202

1 D

ownl

oade

d by

gue

st o

n A

pril

28, 2

021

Dow

nloa

ded

by g

uest

on

Apr

il 28

, 202

1 D

ownl

oade

d by

gue

st o

n A

pril

28, 2

021

Dow

nloa

ded

by g

uest

on

Apr

il 28

, 202

1 D

ownl

oade

d by

gue

st o

n A

pril

28, 2

021

Dow

nloa

ded

by g

uest

on

Apr

il 28

, 202

1 D

ownl

oade

d by

gue

st o

n A

pril

28, 2

021

Page 2: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

Corrections

CELL BIOLOGYCorrection for ‘‘A distinct pool of phosphatidylinositol 4,5-bisphosphate in caveolae revealed by a nanoscale labeling tech-nique,’’ by Akikazu Fujita, Jinglei Cheng, Kumi Tauchi-Sato,Tadaomi Takenawa, and Toyoshi Fujimoto, which appeared inissue 23, June 9, 2009, of Proc Natl Acad Sci USA (106:9256–9261; first published May 22, 2009; 10.1073/pnas.0900216106).

The authors note that on page 9257, Figure 1 appearedincorrectly. This error does not affect the conclusions of thearticle. The corrected figure and its legend appear below.

CELL BIOLOGYCorrection for ‘‘Reprogramming of murine and human somaticcells using a single polycistronic vector,’’ by Bryce W. Carey,Styliani Markoulaki, Jacob Hanna, Kris Saha, Qing Gao,Maisam Mitalipova, and Rudolf Jaenisch, which appeared inissue 1, January 6, 2009, of Proc Natl Acad Sci USA (106:157–162;first published December 24, 2008; 10.1073/pnas.0811426106).

‘‘The authors inadvertently neglected to state that, at the timeof publication, RJ was an advisor to Fate Therapeutics. Weregret this error.’’

www.pnas.org/cgi/doi/10.1073/pnas.0906359106

CHEMISTRYCorrection for ‘‘Development of aliphatic biodegradable pho-toluminescent polymers,’’ by Jian Yang, Yi Zhang, SantoshGautam, Li Liu, Jagannath Dey, Wei Chen, Ralph P. Mason,Carlos A. Serrano, Kevin A. Schug, and Liping Tang, whichappeared in issue 25, June 23, 2009, of Proc Natl Acad Sci USA(106:10086–10091; first published June 8, 2009; 10.1073/pnas.0900004106).

The authors note that due to a printer’s error, Fig. 3B appearedincorrectly. The corrected figure and its legend appear below.

Fig. 1. Labeling of the liposome. (A) Outline of the method. Cells wererapidly frozen, freeze-fractured, and evaporated with carbon (C) and plati-num/carbon (Pt/C) in vacuum. The replica of the split membrane was digestedwith SDS to remove noncast molecules and labeled by GST-PH. Both thecytoplasmic and exoplasmic halves of the membrane were examined. (B)Labeling of small unilamellar liposome replicas. Freeze-fracture replicas ofliposomes containing 95 mol % of phosphatidylcholine (PC) and 5 mol % ofphosphatidylinositol or a phosphoinositide were labeled. Only liposomescontaining PI(4,5)P2 were labeled intensely by GST-PH. A PH mutant, GST-PH(K30N, K32N), which does not bind PI(4,5)P2, showed little labeling in thePI(4,5)P2-containing liposome. (C) Quantification of the GST-PH labeling in theliposomes. The number of gold particles per 1 �m2 of the liposome surface isshown (blue). The labeling on the convex (green) and concave (yellow) sur-faces showed equivalent results.

www.pnas.org/cgi/doi/10.1073/pnas.0906215106

0 2 4 6 8 10 12 14 16 180

20

40

60

80

100

BPLP-Cys 0.2

Rem

ain

ing

Mas

s (%

)

Degradation Time (day)

BPLP-Cys 0.6

A

0 4 8 12 16 20 24 28 32 36

0

20

40

60

80

100

CBPLP-Cys 0.8

Rem

ain

ing

mas

s (%

)

Degradation time (weeks)

CBPLP-Cys 0.2

B

C

0

2

4

6

8

CBPLP-Cys 0.8

CBPLP-Cys 0.6

CBPLP-Cys 0.4

CBPLP-Cys 0.2

Ten

sile

str

ess

(MP

a)

Tensile Strength Young's Modulus

0

50

100

150

200

250

300

CBPLP-Cys 0.8

CBPLP-Cys 0.6

CBPLP-Cys 0.4

CBPLP-Cys 0.2

Elo

ng

atio

n (

%)

D

Fig. 3. Studies of polymer degradation and mechanical properties. (A) Invitro degradation of BPLP-Cys in PBS (pH � 7.4) at 37 °C (n � 5). (B) In vitrodegradation of CBPLP-Cys in PBS (pH � 7.4) at 37 °C (n � 5). (C) Tensile strengthand initial Young’s modulus of CBPLP-Cys synthesized with various molarconcentration of L-cysteine (n � 5). (D) Elongation of CBPLP-Cys synthesizedwith various molar concentration of L-cysteine (n � 5).

www.pnas.org/cgi/doi/10.1073/pnas.0906520106

11818–11819 � PNAS � July 14, 2009 � vol. 106 � no. 28 www.pnas.org

Page 3: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

Reprogramming of murine and human somatic cellsusing a single polycistronic vectorBryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa, Maisam Mitalipovaa,1,and Rudolf Jaenischa,b,1

aWhitehead Institute for Biomedical Research and bDepartment of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142

Contributed by Rudolf Jaenisch, November 11, 2008 (sent for review October 18, 2008)

Directed reprogramming of somatic cells by defined factors pro-vides a novel method for the generation of patient-specific stemcells with the potential to bypass both the practical and ethicalconcerns associated with somatic cell nuclear transfer (SCNT) andhuman embryonic stem (hES) cells. Although the generation ofinduced pluripotent stem (iPS) cells has proven a robust technologyin mouse and human, a major impediment to the use of iPS cells fortherapeutic purposes has been the viral-based delivery of thereprogramming factors because multiple proviral integrationspose the danger of insertional mutagenesis. Here we report a novelapproach to reduce the number of viruses necessary to reprogramsomatic cells by delivering reprogramming factors in a single virususing 2A ‘‘self-cleaving’’ peptides, which support efficient polycis-tronic expression from a single promoter. We find that up to fourreprogramming factors (Oct4, Sox2, Klf4, and c-Myc) can be ex-pressed from a single virus to generate iPS cells in both embryonicand adult somatic mouse cells and we show that a single proviralcopy is sufficient to generate iPS cells from mouse embryonicfibroblasts. In addition we have generated human induced pluri-potent stem (hiPS) cell lines from human keratinocytes, demon-strating that a single polycistronic virus can reprogram humansomatic cells.

2A peptide � four-factor reprogramming � iPS cell � polycistronic

V iral vector-mediated transduction of defined factors hasbeen used to generate induced pluripotent stem (iPS) cells

from embryonic or adult somatic cells in both mouse and human.By gene expression and developmental potential iPS cells areequivalent to embryonic stem (ES) cells maintaining a fullcapacity for differentiation with the ability to form teratomas,generate chimeras, and contribute to the germline (1–8). Thistechnology can be readily applied to many cell types in additionto fibroblasts as numerous cell types have been shown to beamenable to direct reprogramming including pancreatic � cells,neural precursors, and terminally differentiated B cells (9–11).Without the ethical or practical concerns associated with humanembryonic stem cells (hESC), iPS cell technology has emergedas the most promising method for cell-based therapies of regen-erative medicine (12, 13). Yet the current protocols involveviral-based delivery of reprogramming factors leading to multi-ple proviral copies throughout the genome in addition to po-tentially harmful oncogenes, warranting serious concerns re-garding insertional mutagenesis or potential reactivation ofsilenced viral transcripts in cells derived from iPS cell lines.

One improvement to the current strategy would be thedelivery of reprogramming factors within the context of a singlepolycistronic vector. The use of internal ribosomal entry sites(IRES) in polycistronic vectors has been shown to expressmultiple genes from one promoter but this frequently leads tononstoichiometric expression and significantly lower levels of thedownstream cistron(s). One alternative to this method is auton-omous ‘‘self-cleaving’’ 2A peptides originally identified andcharacterized in apthovirus foot-and-mouth disease virus(FMDV) (14, 15). Generally �18–22 aa long, 2A oligopeptidescontain a highly conserved c-terminal D(V/I)EXNPGP motif

that mediates ‘‘ribosomal skipping’’ at the terminal 2A prolineand subsequent amino acid ‘‘2B’’ glycine (16). The most well-characterized 2A peptides are derived from FMDV (hereinreferred to as ‘‘F2A’’), equine rhinitis A virus (ERAV, ‘‘E2A’’),porcine teschovirus-1 (PTV-1, ‘‘P2A’’), and insect Thosea asignavirus (TaV, ‘‘T2A’’). Importantly the ability to express fourproteins in vivo has been achieved using 2A peptides (17).

In this work we examined whether polycistronic vectors using2A peptides can be applied to in vitro reprogramming with thegoal of delivering the reprogramming factors in a single poly-cistronic virus. To test this we developed a four-factor 2A (4F2A)doxycycline (DOX)-inducible lentivirus encoding mouse cDNAsfor Oct4, Sox2, Klf4, and c-Myc separated by three different 2Apeptides (P2A, T2A, and E2A, respectively). Our findings arethreefold: First, we demonstrate that up to three 2A peptidesallow expression of the four reprogramming factors in a singlevector being sufficient to reprogram both embryonic and adultmurine somatic cells. Second, we find that a single proviral copyof the 4F2A is sufficient to reprogram mouse embryonic fibro-blasts (MEFs), suggesting a single transgene could potentiallyreprogram somatic cells. Finally, we show that the 4F2A can beused in reprogramming human cells by generating several iPSlines from neonatal foreskin keratinocytes.

ResultsGeneration of Mouse Ips Cells Using a Single Polycistronic Virus. Ourgoal was to generate polycistronic viral vectors that wouldexpress multiple reprogramming genes from a single promoterusing 2A peptides. For this to occur, one, two, or three 2Aoligopeptides containing unique restriction sites were ligatedinto FUW lentivirus (18) backbones to allow efficient cloning ofOct4, Sox2, c-Myc, and Klf4 each separated by a different 2Asequence. Vectors carrying four, three, or two factors consecu-tively with different combinations of F2A, T2A, E2A, or P2Asequences (Fig. 1A) were tested for their ability to expressindividual factors by transient transfection in human 293 cells.Western blot analysis demonstrated that 2A peptides supportefficient expression of two, three, or all four cistrons from asingle polycistronic vector (Fig. 1B).

A tetracycline inducible lentivirus vector was constructedwhere expression of the genes was controlled from the tetracy-cline operator minimal promoter (tetOP) (Fig. 1C). To testwhether all four genes of a single four-factor (Oct4/Sox2/Klf4/c-Myc) virus could be expressed upon DOX addition, MEFswere infected with the polycistronic vector (referred to below as‘‘4F2A’’) and a constitutive FUW lentivirus carrying the tetra-cycline controllable transactivator (M2rtTA; abbreviated as

Author contributions: B.W.C. and R.J. designed research; B.W.C., S.M., J.H., K.S., Q.G., andM.M. performed research; B.W.C. analyzed data; and B.W.C. and R.J. wrote the paper.

Conflict of interest statement: R.J. is an advisor to Stemgent.

1To whom correspondence may be addressed. E-mail: [email protected] [email protected].

This article contains supporting information online at www.pnas.org/cgi/content/full/0811426106/DCSupplemental.

© 2008 by The National Academy of Sciences of the USA

www.pnas.org�cgi�doi�10.1073�pnas.0811426106 PNAS � January 6, 2009 � vol. 106 � no. 1 � 157–162

CELL

BIO

LOG

Y

Page 4: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

rtTA). Two independent experiments were performed and druginducible expression of the virus was tested 3 days postinfectionby qRT-PCR. Using primers for viral specific transcripts (E2A-cMyc), robust induction was observed (7- to 10-fold) in cellscultured with DOX as compared to control medium (Fig. 1D).To test the relative induction compared to ES cells, Oct4 andSox2 primers that cannot discriminate between viral or endog-enous transcripts were used and in both experiments infectedDOX-induced MEFs were significantly higher than in ES cells(�3.5- and �17-fold over ES levels, respectively). Western blotanalysis of cells isolated at 3 days after infection demonstratedthat little or no protein was expressed when the cells werecultured without DOX whereas robust induction was seen in thepresence of DOX with levels of Oct4 and Sox2 protein beingsimilar to that in ES cells (Fig. 1E).

To test whether the 4F2A vector was able to reprogramsomatic cells to a pluripotent state, MEFs containing a GFPreporter driven by the endogenous Nanog promoter were in-

fected with virus (4F2A � rtTA). Eighty-five to 90% of the cellsstained for Oct4 at 48 h after transduction indicated high titerinfection (Fig. 2A). Morphological changes were observed a fewdays after addition of DOX (data not shown) with distinctcolonies appearing after ?8 days and Nanog-GFP� cells at �25days after DOX induction (Fig. 2B). After mechanical isolationand subsequent passage, the cells had the typical morphology ofES cells and grew independently of DOX. Four independent4F2A iPS cell lines were established that were positive for thepluripotency markers AP, SSEA1, and Nanog-GFP (Fig. 2C).

To investigate whether adult somatic cells could be repro-grammed using the 4F2A vector, we infected tail-tip fibroblasts(TTFs) from 14-week-old mice with the 4F2A � rtTA vectors.Similar to MEFs, typical morphological changes were observeda few days after addition of DOX media. Colonies appeared at?8 days and continued to expand until they were picked (day 16)on the basis of morphology. After several passages, four stableiPS cell lines were established that stained positive for allpluripotency markers (Nanog, Oct4, SSEA1, and AP) (Fig. 2C).MEF iPS cell lines were injected s.c. into severe combinedimmunodeficiency (SCID) mice and were shown to induceteratomas that contained differentiated cells of all three germlayers (Fig. 3A). Finally, injection of MEF iPS cells (no. 4 in Fig.3) into blastocysts generated postnatal chimeras (Fig. 3B),demonstrating that a single 4F2A polycistronic virus can repro-gram MEFs to a pluripotent state.

To determine the number of proviruses carried in the 4F2AiPS cell lines, DNA was extracted and subjected to Southern blotanalysis using an enzyme that does not cut in the vectorsequences. Using Oct4, Sox2, c-Myc, and Klf4 probes for hy-

A

C

B

D

WB: c-Myc

WB: Sox2

WB: Klf4

WB: Oct4

ES -dox +dox

MEF: 4F2A+rtTA

WB: GAPDH

LTR LTR

LTR LTR

LTR LTR

LTR LTR

P2A T2A E2A

Oct4 Sox2 Klf4 cMyctetoPLTRLTR

4F2A: 8.5 kb

Oct4

Sox2

Klf4

cMyc

E

ES SOKM

SOK

SOSMKOKM

WB: anti-KLF4

WB: anti-Oct4

WB: anti-cMyc

WB: anti-Sox2

Lenti-lox Ubi 2A constucts

Sox2-P2A-Oct4-T2A-Klf4-E2A-c-MycSox2-T2A-Oct4-E2A-Klf4Sox2-F2A-Oct4Sox2-F2A-c-MycKlf4-F2A-Oct4Klf4-F2A-c-Myc

SOKM:SOK:

SO:SM:KO:KM:

Fig. 1. Generation of murine iPS cells using a single 4F2A polycistronic virus.(A) FUW lentivirus constructs tested by transient transfection. In total, four 2Apeptides (F2A, T2A, E2A, and P2A) were used. (B) Transient transfection of 293cells with FUW 2A lentiviruses. Cells were harvested after 48 h and analyzed byWestern blot (WB). Efficient protein expression was observed in all constructstested, indicating four unique 2A peptides support robust protein expression.Note: Sox2 protein is not detected in ES cells because only a short exposure wasused. (C) Schematic of the 4F2A DOX-inducible lentivirus containing threetypes of 2A peptides (P2A, T2A, and E2A). Murine cDNAs for Oct4, Sox2, Klf4,and c-Myc. This particular sequence of factors and 2A peptides is subsequentlyreferred to as ‘‘4F2A.’’ (D) RT-PCR anaylsis of mRNA induction in cells trans-duced with OSKM 4F2A � rtTA for 3 days. Total Oct4 or Sox2 induction wasused to test levels of 4F2A induction relative to ES cells. E2A-cMyc primers wereused to detect viral-specific transcripts. Error bars represent SD of the mean oftriplicate reactions. (E) Western blot analysis of MEFs transduced with 4F2A �rtTA for 3 days. Cells infected with 4F2A DOX-inducible lentivirus � rtTAproduce all four reprogramming factors upon addition of doxycycline (DOX).

IH: Oct4

DAPI

+DOX 48 hrs.

MEF: 4F2A+rtTA

Day 9

Day 12

Day 26

Nanog GFP MEFs: OSKM+rtTA (+Dox)A

C

B

Nanog

IH: SSEA1

BF

4F2A MEF iPS #1

AP IH: Nanog

IH:SSEA1

IH: Oct4

BF

4F2A TTF iPS #17

AP

Fig. 2. 4F2A iPS cells express pluripotency markers. (A) Immunostaining ofOct4 protein indicates high titer infections can be achieved with the 4F2A.MEFs were cultured in DOX media for 2 days after transduction with 4F2A �rtTA. (B) Morphology changes in Nanog-GFP MEFs transduced with 4F2A �rtTA cultured in ES media � DOX. Colonies appeared at �8 days, similar to cellsinfected with single viruses. Nanog GFP� colonies were observed by day 25after DOX media removal at day 20. Two columns show typical coloniesobserved on the plate. (C) 4F2A iPS lines generated from Nanog-GFP MEFs or14-week tail-tip fibroblasts (TTFs) that stain positive for pluripotency markersAP, SSEA1, Oct4 and have reactivated the endogenous Nanog locus (GFP� forMEFs and by immunostaining for TTF).

158 � www.pnas.org�cgi�doi�10.1073�pnas.0811426106 Carey et al.

Page 5: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

bridization, we detected bands of identical molecular weightconfirming that the factor sequences were carried in one pro-virus. The total number of proviruses was between one and threewith iPS cell line no. 4 carrying a single viral insert (Fig. 3C). Oneof two integrations from iPS cell line no. 1 failed to produce aband after c-Myc hybridization, suggesting a 3� deletion of thec-Myc sequences may have occurred. A second digest confirmedthe proviral copy numbers (supporting information (SI) Fig.S1A).

To estimate reprogramming efficiency MEFs were infectedwith the 4F2A and rtTA vectors and plated at 0.25 � 106 per10-cm-plate culture dish. About 70% of the MEFs were infectedas estimated by immunostaining of Oct4 at 48 h after infection(Fig. S2A). Cells were cultured in ES media containing DOX for20 days and subsequently transferred to ES cell medium untilGFP� colonies were counted on day 25. An average of �14.7 �4 colonies were detected in three independent dishes (10 � 10� 17) indicating a relative efficiency of 0.0001%. This is twoorders of magnitude lower than that of ‘‘secondary’’ fibroblastsor B cells carrying preselected DOX-inducible proviruses (19).

To test the kinetics of reprogramming using the 4F2A virus we

performed DOX-withdrawl experiments where at specified days(i.e., 2, 4, 8, 12, etc.) DOX-containing media is replaced with ESmedia and the number of Nanog-GFP� colonies are counted atday 25. Using separate drug-inducible viruses to deliver the fourfactors, it has been reported that �9–12 days is the minimumtime required for the generation of stable iPS cells (20, 21). Cellsare not passaged during this time to minimize duplication ofreprogramming events. Two independent experiments wereperformed and in both cases single Nanog-GFP� colonies werepresent on plates cultured in DOX media for 8 days, similar tothe minimum time required using separate viruses (Fig. S1B).

These data demonstrate that a single polycistronic viruscontaining the four factors linked by three 2A peptides allowsfactor expression sufficient to generate iPS cells from embryonicor adult somatic cells. Importantly, our results also show that asingle polycistronic proviral copy is sufficient to reprogramsomatic cells to pluripotency.

Generation of Human Ips Cells Using a Single Polycistronic Virus. Toinvestigate whether human cells could be reprogrammed withthe polycistronic vector, neonatal human foreskin keratinocytes(NHFK) were transduced with both the constitutive rtTA andDOX-inducible 4F2A vectors. The fraction of infected cells was10% as determined by staining for Oct4 at 48 h after transduc-tion (Fig. S3A). Cells were incubated in keratinocyte medium �DOX and allowed to grow for 6 days until they were passagedand cultured in hESC media � DOX on gelatinized plates.Colonies were first detected at day 12 and most displayedtransformed morphology with a few colonies exhibiting a distinctappearance that resembled hESC-like morphology. Two suchcolonies generated in independent infections were picked be-tween 22 and 35 days after infection and found to expand asdistinct colonies with morphology similar to hESC (Fig. 4A).These cells were expanded in the absence of DOX and gave riseto a homogenous population identical to hESC (Ker-iPS) afteran additional two to five passages. The cells stained for thepluripotency markers AP, Oct4, Nanog, Sox2, SSEA4, Tra1–60,and Tra1–81 (Fig. 4B, Fig. S3B) and had a normal karyotype(Fig. 4C). DNA fingerprinting excluded that such Ker-iPS celllines were contamination from previously established human iPScells or hES lines from our lab (data not shown). To determineproviral copy number in Ker-iPS cell lines genomic DNA wasextracted and subjected to Southern blot analysis using anenzyme that does not cut in the vector sequences. Probes for allfour reprogramming factors show hybridization to similar mo-lecular weight band(s) again indicating they were carried on asingle virus. Two different digests (XbaI and BamHI) show the4F2A proviral copy number is three (no. 1.1) and two (no. 3),respectively (Fig. S4 A and B).

To test for pluripotency, one line, Ker-iPS no. 1.1, was injecteds.c. into SCID mice. These cells induced teratomas and afterhistological examination differentiated into cells of all threegerm layers (Fig. 4D). In addition, Ker-iPS no. 1.1 cells, whensubjected to an in vitro neural differentiation protocol, producednestin� neural progenitor cell populations and Tuj1� postmi-totic neurons as detected by immunostaining. (Fig. 4E).

DiscussionThe experiments described in this paper show that up to fourdifferent reprogramming factors inserted into a polycistronicvector separated by 2A sequences can be expressed at levelssufficient to achieve reprogramming. This is in contrast toIRES-based polycistronic vectors where the downstream genesare often expressed at substantially lower levels. Embryonic andadult murine fibroblasts and postnatal human keratinocyteswere induced to form pluripotent iPS cells when infected withthe FUW rtTA and 2A vector transducing Oct4, Sox2, Klf4, andc-Myc. Our finding that a single proviral copy of a polycistronic

23

9

64

2

Kb

Probe: c-Myc

*

#1 #2 #3 #4co

ntrol

Probe: Sox2

*

#1 #2#3 #4co

ntrol

Probe: Klf4

contro

l#1 #2 #3 #4

*

#1 #2#3 #4co

ntrol

*

Probe: Oct4

Proviral copy # 2 3 2 1 2 3 2 1 2 32 1 2 32 1

4F2A iPS#4

iPS#4

Stratified squamous epitheliumSkeletal muscleCiliated epithelium

EctodermMesodermEndoderm

iPS#2

Exocrine glands Skeletal muscle Stratified squamous epithelium

iPS#1

Stratified squamous epitheliumSkeletal muscleCiliated epithelium

A

B

C

4F2A iPS#4

3-weeks 2.5 mo

Fig. 3. 4F2A iPS cells are pluripotent and contain between 1 and 3 proviralintegrations. (A) In vivo differentiation of 4F2A MEF-iPS lines nos. 1, 2, and 4.Histological analysis of teratomas induced after s.c. injection into SCID miceindicates iPS lines contribute to all three germ layers. (B) Moderate-to-highcontribution in postnatal chimeric mice as detected by agouti coat color from4F2A iPS line no. 4. (C) Southern blot analysis of 4F2A proviral integrations inMEF-iPS cell line nos. 1–4. iPS cell DNA was digested with BamHI. Hybridizationof the same molecular weight fragment using all four probes indicates pres-ence of 4F2A provirus. Red arrow highlights iPS line no. 4, which contained oneproviral copy of the 4F2A. * indicates endogenous allele.

Carey et al. PNAS � January 6, 2009 � vol. 106 � no. 1 � 159

CELL

BIO

LOG

Y

Page 6: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

vector suggests that with sufficient expression it may be possibleto reprogram using a stably expressed transgene. This is sup-ported by the reports suggesting insertional mutagenesis is notnecessary for reprogramming and that transient expression ofreprogramming factors is sufficient to generate iPS cells (22, 23).Preliminary work by Okita et al. (2008) achieved reprogrammingof murine embryonic fibroblasts using Moloney viruses express-ing three factors separated by F2A peptides but with a significantreduction in efficiency. We extend these findings by demonstrat-ing that even at a low efficiency, 2A vectors can reprogram adultmurine somatic cells and human keratinocytes.

In addition, Okita et al. (2008) were able to reprogram murineembryonic fibroblasts using multiple transient transfections oftwo plasmids expressing Oct4-Klf4-Sox2 and c-Myc, respectively.Although iPS cells were generated with greatly reduced effi-ciency, iPS cell lines with no detectable vector integration wererecovered. Interestingly, many of the iPS cell lines generatedafter transient transfection contained integrated plasmids, sug-gesting that stable integration of exogenous sequences can occur.

We observe a reprogramming efficiency significantly lowerthan previous experiments using single vectors to transduce eachof the four factors (Fig. S2B and Table S1). It is possible that thelower reprogramming efficiency is because of the stochiometryof factor expression from the polycistronic vector, which may besuboptimal for inducing reprogramming. Transduction withseparate vectors allows integration of different numbers ofproviruses for each factor, therefore reprogramming may selectfor a specific set of proviral integrations that result in highexpression or an optimal stochiometry between the differentfactors. This may not be possible using the 2A system, which hasbeen reported to support near equimolar protein expression invivo (17). Also, when separate vectors transducing each of thefour factors were used for induction of iPS cells, Nanog-GFP�cells were detected as early as 16 days after DOX induction incontrast to GFP� cells observed 22–25 days after 4F2A vectortransduction, consistent with less optimal reprogramming.Moreover, whereas iPS cells frequently carry multiple Oct4 orKlf4 proviruses, consistently fewer Sox2 proviruses were found,suggesting that a high level of Sox2 expression may be unfavor-able for reprogramming (24).

Ultimately multiple technologies may converge to allow thegeneration of therapeutically acceptable iPS cells without viralintegrations or lingering reprogramming factors within the ge-nome. In a recent proof-of-principle work nonintegrating ad-enoviruses were used alone or in tandem with an inducible Oct4transgene to reprogram mouse hepatocytes and fibroblasts,respectively, demonstrating insertional mutagenesis is not nec-cessary for reprogramming (23). iPS cells without detectableviral integrations were recovered; however, efficiencies weresignificantly lower than Moloney or lentivirus reprogramming offibroblasts. It will be important to determine whether other moreaccessible cell types are amenable to adenovirus-mediated re-programming as hepatocytes are harder to obtain and grow fromhuman patients in comparison to keratinocytes or fibroblasts.Furthermore transient technologies such as adenovirus or DNAtransfection of reprogramming factors may not have the abilityto express reprogramming factors throughout the minimum timeinterval at levels necessary for acquisition of an iPS state,especially important for human reprogramming. Because thefour factors are expressed from a defined location, the polycis-tronic vector system described in this paper should simplify thestudy of reprogramming mechanisms and may facilitate theexcision of the vector resulting in iPS cells that carry noexogenous transgenes.

Materials and MethodsViral Preparation and Infection. Construction of 4F2A lentiviral vectors con-taining Oct4, Sox2, Klf4, and c-Myc under control of the tetracycline operatorand a minimal CMV promoter was generated after EcoRI cloning from a FUWlentivirus backbone. All constructs were generated using unique restrictionsites after amplification by PCR to place an individual factor between arespective 2A peptide (first, XbaI-NheI; second, SphI; third, XhoI; and fourth,AscI). Respective 2A sequences: P2A, GCCACGAAGCAAGCAGGAGATGTTGAA-GAAAACCCCGG GCCT; T2A, GAGGGCAGAGGAAGTCTTCTAACATGCG-GTGACGTGGAGGAGA ATCCCGGCCCT; and E2A, CAGTGTACTAATTAT-GCTCTCTTGAAATTGGCTGG AGATGTTGAGAGCAACCCAGGTCCC).Replication-incompetent lentiviral particles (4F2A and M2rtTA) were pack-aged in 293T cells with a VSV-G coat and used to infect MEFs containing a GFPallele targeted to the endogenous Nanog locus (7, 25). Fourteen-week-oldtail-tip fibroblasts were derived from mice previously published (12). Humankeratinocytes (NHFK) were obtained from Coriell Institute for Medical Re-search. Viral supernatants from cultures packaging each of the two viruseswere pooled, filtered through a 0.45-�M filter and subjected to ultracentrif-ugation for concentration. Virus pellets were resuspended in ES cell medium(DMEM supplemented with 10% FBS (HyClone), leukemia inhibitory factor,�-mercaptoethanol (Sigma-Aldrich), penicillin/streptomycin, L-glutamine,and nonessential amino acids (all from Invitrogen) before being applied tocells for 24 h.

p3 Ker #1.1 p5 Ker #1.1 (no dox)

KeratinocyteshiPS Ker #1.1A

IH: Oct-4 IH: AP

IH: Tra1-60IH: Tra1-60IH: SSEA-4 IH: Tra1-81

IH: Nanog

B C

D

E

MesodermEctodermEndoderm

Intestine-like epithelium Neural rosettes Cartilage

IH: Tuj-1 DAPI IH: Tuj-1IH: Nestin DAPI

hiPS Ker #1.1hiPS Ker #1.1

Fig. 4. Generation of human iPS lines using a single 4F2A polycistronicvirus. (A) Neonatal human foreskin keritinocytes (NHFK) transduced with4F2A (carrying mouse cDNAs) � rtTA. On day 22 a single colony was pickedand expanded, giving rise to colonies resembling hES colonies. Thesecolonies were picked and a stable hiPS line was established. (B) Ker-hiPS no.1.1 immunostaining for pluripotency markers AP, Oct4, Nanog, SSEA-4,Tra1– 60, and Tra1– 81. DAPI stain is in lower panels. (C) Karyotype ofKer-hiPS no. 1.1 is normal 46 XY. (D) In vivo differentiation of Ker-hiPS no.1.1. Hematoxylin and eosin staining of teratoma sections generated byKer-hiPS no. 1.1. (E) In vitro differentiation of Ker-hiPS no. 1.1. (Left) Ker-iPSno. 1.1-derived neural precursors exposed to differentiation conditions for6 days produce terminally differentiated neurons as detected by anti-Tuj1immunostaining (green). (Right) Ker-iPS no. 1.1 neural precursors (NPs)undergo spontaneous differentiation. NPs were detected by anti-Nestinimmunostaining and differentiated neurons by anti-Tuj1 (red). DAPI stainfor DNA in both pictures is blue.

160 � www.pnas.org�cgi�doi�10.1073�pnas.0811426106 Carey et al.

Page 7: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

Western Blot. One hundred microliters of lysis buffer containing 2% SDS, 10mM DTT, 10% glycerol, 12% urea, 10 mM Tris-HCl (pH 7.5), 1 mM phenyl-methylsulfonyl fluoride, 1� protease inhibitor mixture (Roche), 25 �MMG132 proteosome inhibitor, and boiled for 5 min. Proteins were thenquantified using Bradford reagent (Pierce) and taking spectrophotometricreadings at 590 nm. Concentrations were estimated against a standardcurve generated using BSA. Total protein (5 �g) was subjected to electro-phoreses in a denaturing 10% polyacrylamide gel containing 10% SDS.Proteins were then transferred onto Immobilon-P membranes (Millipore)using a semi-dry transfer apparatus. Membranes were blocked in PBS,0.01% Tween 20 containing 2% nonfat powdered milk (Bio-Rad). Proteinswere detected by incubating with antibodies at a concentration of 50ng/mL in blocking solution. Antibodies used were Oct4 (h-134, Santa CruzBiotechnology); Sox2 (mouse monoclonal, R&D Biosystems); c-Myc (06 –340, Upstate); Klf4 (H-180, Santa Cruz Biotechnology); GAPDH (sc-25778,Santa Cruz Biotechnology).

Quantitative RT-PCR. Total RNA was isolated using TRIzol reagent (Invitrogen).Five micrograms of total RNA was treated with DNase I to remove potentialcontamination of genomic DNA using a DNA Free RNA kit (Zymo Research).One microgram of DNase I-treated RNA was reverse transcribed using a FirstStrand Synthesis kit (Invitrogen) and ultimately resuspended in 100 �L ofwater. Quantitative PCR analysis was performed in triplicate using 1/50 of thereverse transcription reaction in an ABI Prism 7000 (Applied Biosystems) withPlatinum SYBR green qPCR SuperMix-UDG with ROX (Invitrogen). Equal load-ing was achieved by amplifying GAPDH mRNA and all reactions were per-formed in triplicate. Primers used for amplification were as follows:

Oct4 F, 5�-ACATCGCCAATCAGCTTGG-3� andR, 5�-AGAACCATACTCGAACCACATCC-3�Sox2 F, 5�-ACAGATGCAACCGATGCACC-3� andR, 5�- TGGAGTTGTACTGCAGGGCG-3�4F2A (E2A-cMyc) F, 5�-GGCTGGAGATGTTGAGAGCAA-3� andR, 5�-AAAGGAAATCCAGTGGCGCGAPDH F, 5�-TTCACCACCATGGAGAAGGC-3� andR, 5�-CCCTTTTGGCTCCACCCT-3�.Error bars represent SD of the mean of triplicate reactions.

Southern Blotting. Ten micrograms of BamHI digested genomic DNA wereseparated on a 0.7% agarose gel, transferred to a nylon membrane (Amer-sham), and hybridized with 32P random primer (Stratagene) labeled probes forOCT4 (EcoRI-PstI fragment of pFUW-tetO-OCT4 plasmid), KLF4 (full-lengthKLF4 cDNA), c-MYC (full-length c-MYC cDNA), and SOX2 (full-length fragmentof pFUW-tetO-SOX2 plasmid).

Immunofluorescent Staining. Cells were fixed in 4% paraformaldehyde for 20min at 25 °C, washed three times with PBS, and blocked for 15 min with 5% FBSin PBS containing 0.1% Triton-X. After incubation with primary antibodiesagainst Oct4 (Santa Cruz, h-134); Sox2 (R&D Biosystems); Nanog (anti-ms andanti-h, R&D Biosystems); Tra-1–60 (mouse monoclonal, Chemicon Interna-tional); hNANOG (goat polyclonal, R&D Bioystems); mNANOG (A300–398A,Bethyl Laboratories); Tra1–81 (mouse monoclonal, Chemicon International);and SSEA4 and SSEA1 (monoclonal mouse, Developmental Studies HybridomaBank) for 1 h in 1% FBS in PBS containing 0.1% Triton-X, cells were washed

three times with PBS and incubated with fluorophore-labeled appropriatesecondary antibodies purchased from Jackson Immunoresearch. Specimenswere analyzed on an Olympus fluorescence microscope and images wereacquired with a Zeiss Axiocam camera.

Mouse Chimera and Teratoma Formation. Diploid blastocysts (94�98 h afterhCG injection) were placed in a drop of Hepes-CZB medium under mineraloil. A flat tip microinjection pipette with an internal diameter of 16 �m wasused for iPS cell injections. Each blastocyst received 8 –10 iPS cells. Afterinjection, blastocysts were cultured in potassium simplex optimizationmedium (KSOM) and placed at 37 °C until transferred to recipient females.About 10 injected blastocysts were transferred to each uterine horn of2.5-day-postcoitum pseudopregnant B6D2F1 female. Pups were recoveredat day 19.5 and fostered to lactating B6D2F1 mothers when necessary.Teratoma formation was performed by depositing 2 � 106 cells under theflanks of recipient SCID or Rag2�/� mice. Tumors were isolated 3– 6 weekslater for histological analysis.

Human Teratoma Formation and Analysis. hiPSCs were collected by collage-nase treatment (1.5 mg/mL) and separated from feeder cells by subsequentwashes with medium and sedimentation of iPSC colonies. iPSC aggregateswere collected by centrifugation and resuspended in a ratio of 106 cells in250 �L of iPSC culture media. iPSCs were injected s.c. by a 21-gauge needlein the back of SCID mice (Taconic). A tumor developed within 6 weeks andthe animal was killed before tumor size exceeded 1.5 cm in diameter.Teratomas were isolated after killing the mice and fixed in formalin. Aftersectioning, teratomas were diagnosed on the basis of hematoxylin andeosin staining. Karyotype analysis was done with Cell Line Genetics.

In Vitro Differentiation of Human IPS Cells into Neuronal Progenitors. Humankeratinocyte iPS cells were allowed to outgrow in culture without pasagingfor 2 weeks with daily medium change. At day 15 after passage, distinct neuralrossets were observed and picked mechanically by pooled glass pipett (26).Rosettes were replated on dishes precoated with 15 �g/mL polyornithin/10�g/mL of laminin (Po/Lam) in N2B27 medium supplemented with FGF2 (20ng/mL) EGF (20 ng/mL) (all R&D Biosystems). After 5–7 days, cells were disso-ciated by scraping with cell lifter and pipetting to single cells in N2B27 mediumand replated to Po/Lam culture dishes.

Differentiation and Immunocytochemistry. Induction of differentiation of neu-ral progenitors was performed by withdrawal of FGF2 and EGF from culturemedium for 5 days. Cells were fixed in 4% paraformaldehyde for 20 min andstained for human nestin (Chemicon; 1:100) and Tuj-1 (1:100) and subse-quently washed three times with PBS and incubated with fluorophore-labeledappropriate secondary antibodies purchased from Jackson Immunoresearch.Specimens were analyzed on an Olympus fluorescence microscope and imageswere acquired with a Zeiss Axiocam camera.

ACKNOWLEDGMENTS. We thank D. Hockemeyer and F. Soldner for the FUWM2rtTA (27) and M. Wernig, C. Lengner, M. Creyghton, R. Flannery, J. Daus-man, D. Fu, and members of the Jaenisch lab for excellent assistance andhelpful comments. R.J. is supported by grants from the National Institutes ofHealth: 5-RO1-HDO45022, 5-R37-CA084198, and 5-RO1-CA087869. J.H. is aNovartis Fellow by the Helen Hay Whitney Foundation.

1. Lowry WE, et al. (2008) Generation of human induced pluripotent stem cells fromdermal fibroblasts. Proc Natl Acad Sci USA 105:2883–2888.

2. Maherali N, et al. (2007) Directly reprogrammed fibroblasts show global epigeneticremodeling and widespread tissue contribution. Cell Stem Cell 1:55–70.

3. Okita K, Ichisaka T, Yamanaka S (2007) Generation of germline-competent inducedpluripotent stem cells. Nature 448:313–317.

4. Park IH, et al. (2008) Reprogramming of human somatic cells to pluripotency withdefined factors. Nature 451:141–146.

5. Takahashi K, et al. (2007) Induction of pluripotent stem cells from adult humanfibroblasts by defined factors. Cell 131:861–872.

6. Takahashi K, Yamanaka S (2006) Induction of pluripotent stem cells from mouseembryonic and adult fibroblast cultures by defined factors. Cell 126:663–676.

7. Wernig M, et al. (2007) In vitro reprogramming of fibroblasts into a pluripotentES-cell-like state. Nature 448:318–324.

8. Yu J, et al. Induced pluripotent stem cell lines derived from human somatic cells (2007)Science 318:1917–1920.

9. Hanna J, et al. (2008) Direct reprogramming of terminally differentiated mature Blymphocytes to pluripotency. Cell 133:250–264.

10. Kim JB, et al. (2008) Pluripotent stem cells induced from adult neural stem cells byreprogramming with two factors. Nature 454:646–650.

11. Stadtfeld M, Brennand K, Hochedlinger K (2008) Reprogramming of pancreatic betacells into induced pluripotent stem cells. Curr Biol 18:890–894.

12. Hanna J, et al. (2007) Treatment of sickle cell anemia mouse model with iPS cellsgenerated from autologous skin. Science 318:1920–1923.

13. Wernig M, et al. (2008) Neurons derived from reprogrammed fibroblasts functionallyintegrate into the fetal brain and improve symptoms of rats with Parkinson’s disease.Proc Natl Acad Sci USA 105:5856–5861.

14. Ryan MD, Drew J (1994) Foot-and-mouth disease virus 2A oligopeptide mediatedcleavage of an artificial polyprotein. EMBO J 13:928–933.

15. Ryan MD, King AM, Thomas GP (1991) Cleavage of foot-and-mouth disease viruspolyprotein is mediated by residues located within a 19 amino acid sequence J GenVirol 72(Pt 11), 2727–2732.

16. Doronina VA, et al. (2008) Site-specific release of nascent chains from ribosomes at asense codon. Mol Cell Biol 28:4227–4239.

17. Szymczak AL, et al. (2004) Correction of multi-gene deficiency in vivo using a single‘self-cleaving’ 2A peptide-based retroviral vector. Nat Biotechnol 22:589–594.

18. Lois C, et al. (2002) Germline transmission and tissue-specific expression of transgenesdelivered by lentiviral vectors. Science 295:868–872.

19. Wernig M, et al. (2008) A drug-inducible transgenic system for direct reprogrammingof multiple somatic cell types. Nat Biotechnol 26:916–924.

20. Brambrink T, et al. (2008) Sequential expression of pluripotency markers during directreprogramming of mouse somatic cells. Cell Stem Cell 2:151–159.

21. Stadtfeld M, et al. (2008) Defining molecular cornerstones during fibroblast to iPS cellreprogramming in mouse. Cell Stem Cell 2:230–240.

Carey et al. PNAS � January 6, 2009 � vol. 106 � no. 1 � 161

CELL

BIO

LOG

Y

Page 8: Proc Natl Acad Sci USAReprogramming of murine and human somatic cells using a single polycistronic vector Bryce W. Careya,b, Styliani Markoulakia, Jacob Hannaa, Kris Sahaa, Qing Gaoa,

22. Okita K, et al. (2008) Generation of mouse induced pluripotent stem cells without viralvectors. Science 322:949–953.

23. Stadtfeld M, et al. (2008) Induced pluripotent stem cells generated without viralintegration. Science 322:945–949.

24. Eminli S, Utikal JS, Arnold K, Jaenisch R, Hochedlinger K (2008) Reprogramming ofneural progenitor cells into iPS cells in the absence of exogenous Sox2 expression. StemCells 26:2467–2474.

25. Meissner A, Wernig M, Jaenisch R (2007) Direct reprogramming of geneti-cally unmodified fibroblasts into pluripotent stem cells. Nat Biotechnol 25:1177–1181.

26. Zhang SC, et al. (2001) In vitro differentiation of transplantable neural precursors fromhuman embryonic stem cells. Nat Biotechnol 19:1129–1133.

27. Hockemeyer D, et al. (2008) A drug-inducible system for direct reprogramming ofhuman somatic cells to pluripotency. Cell Stem Cell 3:346–353.

162 � www.pnas.org�cgi�doi�10.1073�pnas.0811426106 Carey et al.