Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C)...

6
Differentiation-defective phenotypes revealed by large-scale analyses of human pluripotent stem cells Michiyo Koyanagi-Aoi a,1 , Mari Ohnuki a,1 , Kazutoshi Takahashi a , Keisuke Okita a , Hisashi Noma b , Yuka Sawamura a , Ito Teramoto a , Megumi Narita a , Yoshiko Sato a , Tomoko Ichisaka a , Naoki Amano a , Akira Watanabe a , Asuka Morizane a , Yasuhiro Yamada a,c , Tosiya Sato d , Jun Takahashi a,e , and Shinya Yamanaka a,f,2 a Center for iPS Cell Research and Application, e Department of Biological Repair, Institute for Frontier Medical Sciences, and c Institute for Integrated Cell- Material Sciences, Kyoto University, Kyoto 606-8507, Japan; b Department of Data Science, The Institute of Statistical Mathematics, Tokyo 190-8562, Japan; d Department of Biostatistics, Kyoto University School of Public Health, Kyoto 606-8501, Japan; and f Gladstone Institute of Cardiovascular Disease, San Francisco, CA 94158 Contributed by Shinya Yamanaka, October 30, 2013 (sent for review September 13, 2013) We examined the gene expression and DNA methylation of 49 human induced pluripotent stem cells (hiPSCs) and 10 human embryonic stem cells and found overlapped variations in gene expression and DNA methylation in the two types of human pluripotent stem cell lines. Comparisons of the in vitro neural differentiation of 40 hiPSCs and 10 human embryonic stem cells showed that seven hiPSC clones retained a signicant number of undifferentiated cells even after neural differentiation culture and formed teratoma when transplanted into mouse brains. These differentiation-defective hiPSC clones were marked by higher expression levels of several genes, including those expressed from long terminal repeats of specic human endogenous retroviruses. These data demonstrated a subset of hiPSC lines that have aberrant gene expression and defective potential in neural differentiation, which need to be identied and eliminated before applications in regenerative medicine. H uman pluripotent stem cells possess a robust potential for proliferation and provide useful sources of cells for re- generative medicine and drug discovery. Two types of human pluripotent stem cells have been generated: human embryonic stem cells (hESCs) derived from blastocysts (1) and induced pluripotent stem cells (hiPSCs), which are generated from so- matic cells by factor-mediated reprogramming (2, 3). In the past few years, ndings have been controversial in regard to whether hESCs and hiPSCs are distinct cell types. Some reports have argued that they could not be clearly distin- guished (46), whereas others have reported that they have dif- ferences in their gene expression (710), DNA methylation (1013), and capacity for differentiation (14). In the latter papers, relatively small numbers of cell lines were generally compared. In addition, most comparisons used pluripotent cell lines from various laboratories, so the observed differences may be attrib- utable to laboratory-specic variations owing to technical dif- ferences (15). To overcome these problems, we compared the mRNA and microRNA (miRNA) expression and DNA methylation between 10 hESCs and 49 hiPSCs that had been cultured under the same conditions. Furthermore, we compared the in vitro directed neural differentiation of these pluripotent stem cells. Results Overlapped Variations of mRNA Expression and DNA Methylation in hiPSCs and hESCs. We analyzed a total of 49 hiPSCs derived from four types of somatic cells, including human dermal broblasts (HDFs), dental-pulp stem cells (DP), cord blood cells (CB), and peripheral blood mononuclear cells (PBMN), generated using three gene delivery methods, including those using retroviruses, nonintegration episomal plasmids, and Sendai viruses (Table 1 and Dataset S1). Most clones were generated in our own labo- ratory, except for three clones that were established in another laboratory (16). Before the analyses of gene/miRNA expression (Fig. 1 A and B) and DNA methylation (Fig. 1C) we cultured these hiPSCs, as well as 10 hESCs, under the same culture conditions for at least three passages. In addition, we analyzed the original somatic cells, two human embryonic carcinoma cell (hECC) lines (NTera2 cloneD1 and 2102Ep 4D3), and three cancer cell lines (HepG2, MCF7, and Jurkat). The mRNA microarray analyses (Fig. 1A) identied 61 probes with signicant differences in expression between hESCs and hiPSCs [t test, false discovery rate (FDR) <0.05]. Each of the 61 probes showed variable expression among both the hESCs and hiPSCs, and the distributions of the expression levels in the two groups overlapped (Fig. 1D). Of note, hESCs established at Kyoto University (Kyoto hESCs) were more similar to hiPSCs than to the remaining hESCs (other hESCs) in their expression of 15 probes that were differentially expressed between hESCs and hiPSCs [FDR <0.05 and fold change (FC) >3] (Fig. S1A). In contrast, hierarchical clustering using all probes showed no clear- cut separation among Kyoto hESCs, other ESCs, and iPSCs, indicating that the similarities between Kyoto ESCs and iPSCs are conned to a small set of genes (Fig. S1B). In addition, the miRNA array analyses (Fig. 1B) did not nd any signicant differences between hESCs and hiPSCs (t test, FDR <0.05). The expressions of hsa-miR-886-3p and hsa-miR-142-3p tended to be higher in hiPSCs, but the expression levels of these Signicance In the past few years, ndings have been controversial in regard to whether human induced pluripotent stem cells (hiPSCs) are distinct from human embryonic stem cells (hESCs) in their molecular signatures and differentiation properties. In this study, hiPSCs and hESCs have overlapping variations in molecular signatures such as RNA expression and DNA meth- ylation. However, some hiPSC clones retained a signicant number of undifferentiated cells even after neural differenti- ation culture and formed teratoma when transplanted into mouse brains. These differentiation-defective hiPSC clones were marked by higher expression levels of several genes, including those expressed from long terminal repeats of specic human endogenous retroviruses. They need to be identied and elimi- nated prior to applications in regenerative medicine. Author contributions: M.K.-A., M.O., K.T., and S.Y. designed research; M.K.-A., M.O., K.T., K.O., Y. Sawamura, I.T., M.N., Y. Sato, and T.I. performed research; A.M. and J.T. sup- ported the neural differentiation experiments; Y.Y. supported the pathological analysis; H.N. and T.S. supported the statistical analysis; M.K.-A., M.O., H.N., N.A., and A.W. ana- lyzed data; and M.K.-A., M.O., K.T., and S.Y. wrote the paper. Conict of interest statement: S.Y. is a member without salary of the scientic advisory boards of iPierian, iPS Academia Japan, Megakaryon Corporation, and HEALIOS K. K. Japan. Data deposition: Gene expression, miRNA expression, DNA methylation, and exon array data have been deposited in the Gene Expression Omnibus (GEO) database, www.ncbi. nlm.nih.gov/geo (accession no. GSE49053). 1 M.K.-A. and M.O. contributed equally to this work. 2 To whom correspondence should be addressed. E-mail: [email protected]. This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10. 1073/pnas.1319061110/-/DCSupplemental. www.pnas.org/cgi/doi/10.1073/pnas.1319061110 PNAS | December 17, 2013 | vol. 110 | no. 51 | 2056920574 CELL BIOLOGY Downloaded by guest on February 9, 2021

Transcript of Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C)...

Page 1: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

Differentiation-defective phenotypes revealed bylarge-scale analyses of human pluripotent stem cellsMichiyo Koyanagi-Aoia,1, Mari Ohnukia,1, Kazutoshi Takahashia, Keisuke Okitaa, Hisashi Nomab, Yuka Sawamuraa,Ito Teramotoa, Megumi Naritaa, Yoshiko Satoa, Tomoko Ichisakaa, Naoki Amanoa, Akira Watanabea, Asuka Morizanea,Yasuhiro Yamadaa,c, Tosiya Satod, Jun Takahashia,e, and Shinya Yamanakaa,f,2

aCenter for iPS Cell Research and Application, eDepartment of Biological Repair, Institute for Frontier Medical Sciences, and cInstitute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8507, Japan; bDepartment of Data Science, The Institute of Statistical Mathematics, Tokyo 190-8562, Japan;dDepartment of Biostatistics, Kyoto University School of Public Health, Kyoto 606-8501, Japan; and fGladstone Institute of Cardiovascular Disease,San Francisco, CA 94158

Contributed by Shinya Yamanaka, October 30, 2013 (sent for review September 13, 2013)

We examined the gene expression and DNA methylation of 49human induced pluripotent stem cells (hiPSCs) and 10 humanembryonic stem cells and found overlapped variations in geneexpression and DNA methylation in the two types of humanpluripotent stem cell lines. Comparisons of the in vitro neuraldifferentiation of 40 hiPSCs and 10 human embryonic stem cellsshowed that seven hiPSC clones retained a significant number ofundifferentiated cells even after neural differentiation culture andformed teratoma when transplanted into mouse brains. Thesedifferentiation-defective hiPSC clones were marked by higherexpression levels of several genes, including those expressed fromlong terminal repeats of specific human endogenous retroviruses.These data demonstrated a subset of hiPSC lines that have aberrantgene expression and defective potential in neural differentiation,which need to be identified and eliminated before applications inregenerative medicine.

Human pluripotent stem cells possess a robust potential forproliferation and provide useful sources of cells for re-

generative medicine and drug discovery. Two types of humanpluripotent stem cells have been generated: human embryonicstem cells (hESCs) derived from blastocysts (1) and inducedpluripotent stem cells (hiPSCs), which are generated from so-matic cells by factor-mediated reprogramming (2, 3).In the past few years, findings have been controversial in

regard to whether hESCs and hiPSCs are distinct cell types.Some reports have argued that they could not be clearly distin-guished (4–6), whereas others have reported that they have dif-ferences in their gene expression (7–10), DNA methylation (10–13), and capacity for differentiation (14). In the latter papers,relatively small numbers of cell lines were generally compared.In addition, most comparisons used pluripotent cell lines fromvarious laboratories, so the observed differences may be attrib-utable to laboratory-specific variations owing to technical dif-ferences (15).To overcome these problems, we compared the mRNA and

microRNA (miRNA) expression and DNA methylation between10 hESCs and 49 hiPSCs that had been cultured under the sameconditions. Furthermore, we compared the in vitro directedneural differentiation of these pluripotent stem cells.

ResultsOverlapped Variations of mRNA Expression and DNA Methylation inhiPSCs and hESCs. We analyzed a total of 49 hiPSCs derived fromfour types of somatic cells, including human dermal fibroblasts(HDFs), dental-pulp stem cells (DP), cord blood cells (CB), andperipheral blood mononuclear cells (PBMN), generated usingthree gene delivery methods, including those using retroviruses,nonintegration episomal plasmids, and Sendai viruses (Table 1and Dataset S1). Most clones were generated in our own labo-ratory, except for three clones that were established in anotherlaboratory (16). Before the analyses of gene/miRNA expression(Fig. 1 A and B) and DNA methylation (Fig. 1C) we cultured thesehiPSCs, as well as 10 hESCs, under the same culture conditions for

at least three passages. In addition, we analyzed the original somaticcells, two human embryonic carcinoma cell (hECC) lines (NTera2cloneD1 and 2102Ep 4D3), and three cancer cell lines (HepG2,MCF7, and Jurkat).The mRNA microarray analyses (Fig. 1A) identified 61 probes

with significant differences in expression between hESCs andhiPSCs [t test, false discovery rate (FDR) <0.05]. Each of the 61probes showed variable expression among both the hESCs andhiPSCs, and the distributions of the expression levels in the twogroups overlapped (Fig. 1D). Of note, hESCs established atKyoto University (Kyoto hESCs) were more similar to hiPSCsthan to the remaining hESCs (other hESCs) in their expressionof 15 probes that were differentially expressed between hESCsand hiPSCs [FDR <0.05 and fold change (FC) >3] (Fig. S1A). Incontrast, hierarchical clustering using all probes showed no clear-cut separation among Kyoto hESCs, other ESCs, and iPSCs,indicating that the similarities between Kyoto ESCs and iPSCsare confined to a small set of genes (Fig. S1B). In addition, themiRNA array analyses (Fig. 1B) did not find any significantdifferences between hESCs and hiPSCs (t test, FDR <0.05). Theexpressions of hsa-miR-886-3p and hsa-miR-142-3p tendedto be higher in hiPSCs, but the expression levels of these

Significance

In the past few years, findings have been controversial inregard to whether human induced pluripotent stem cells(hiPSCs) are distinct from human embryonic stem cells (hESCs)in their molecular signatures and differentiation properties. Inthis study, hiPSCs and hESCs have overlapping variations inmolecular signatures such as RNA expression and DNA meth-ylation. However, some hiPSC clones retained a significantnumber of undifferentiated cells even after neural differenti-ation culture and formed teratoma when transplanted intomouse brains. These differentiation-defective hiPSC clones weremarked by higher expression levels of several genes, includingthose expressed from long terminal repeats of specific humanendogenous retroviruses. They need to be identified and elimi-nated prior to applications in regenerative medicine.

Author contributions: M.K.-A., M.O., K.T., and S.Y. designed research; M.K.-A., M.O., K.T.,K.O., Y. Sawamura, I.T., M.N., Y. Sato, and T.I. performed research; A.M. and J.T. sup-ported the neural differentiation experiments; Y.Y. supported the pathological analysis;H.N. and T.S. supported the statistical analysis; M.K.-A., M.O., H.N., N.A., and A.W. ana-lyzed data; and M.K.-A., M.O., K.T., and S.Y. wrote the paper.

Conflict of interest statement: S.Y. is a member without salary of the scientific advisoryboards of iPierian, iPS Academia Japan, Megakaryon Corporation, and HEALIOS K. K.Japan.

Data deposition: Gene expression, miRNA expression, DNA methylation, and exon arraydata have been deposited in the Gene Expression Omnibus (GEO) database, www.ncbi.nlm.nih.gov/geo (accession no. GSE49053).1M.K.-A. and M.O. contributed equally to this work.2To whom correspondence should be addressed. E-mail: [email protected].

This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1319061110/-/DCSupplemental.

www.pnas.org/cgi/doi/10.1073/pnas.1319061110 PNAS | December 17, 2013 | vol. 110 | no. 51 | 20569–20574

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1

Page 2: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

miRNAs showed overlapped variations among hiPSCs andhESCs (Fig. S1A).We next compared the global DNA methylation status be-

tween hiPSCs and hESCs by the Illumina Infinium HumanMethylation27 BeadChip assay. Among 27,445 CpG dinucleotides

examined, we did not identify significantly differentially meth-ylated CpG regions (CG-DMR) between the hESCs and hiPSCs(Mann–Whitney U test, FDR <0.05) (Fig. 1C).We then validated the CG-DMRs reported in previous stud-

ies. Three studies identified a total of 205 regions as CG-DMRs,including 130, 71, and 4 regions identified by comparing fivehiPSCs and two hESCs (13), three versus three (12), and nineversus three (10) cell lines, respectively. Of the 205 regions, 46regions containing 66 CpG dinucleotides were covered by theInfinium platform used in our study (Table S1A). Based on themethylation levels in our hiPSCs and hESCs, these CpGs wereclustered into three groups (Fig. 1E). Two-thirds of these CpGsbelonged to group A: They tended to be highly methylated inhESCs, ECCs, and cancer cell lines and to be hypomethylated inhiPSCs, as well as somatic cells. However, they were also hypo-methylated in Kyoto ESCs (17). The methylation status of theupstream region of the paraoxonase 3 (PON3), a representativeexample of CpGs in group A, was confirmed by pyrosequencing(Fig. 1F). Thus, the CpG methylation status in group A may

Table 1. Summary of the iPSC clones used in this study

Method used to generate clones

Origin Retrovirus Episomal plasmid Sendaivirus Total

HDFs 22 (5, 0) 3 (0, 0) 0 (0, 0) 25 (5, 0)DP 1 (0, 0) 2 (1, 1) 0 (0, 0) 3 (1, 1)CB 3 (1, 2) 4 (0, 0) 5 (0, 0) 12 (1, 2)PBMN 0 (0, 0) 5 (0, 1) 4 (0, 0) 9 (0, 1)Total 26 (6, 2) 14 (1, 2) 9 (0, 0) 49 (7, 4)

Total clone numbers with type-1 defective clone numbers and type-2defective clone numbers in parentheses are shown.

A

Baverage of 10 hESCs

aver

age

of 4

9 hi

PS

Cs

mRNA expression

86420-2-4-6

86420

-2-4-6

miRNA expressionhsa-miR-886-3p

hsa-miR-142-3p

50-5-10

5

0

-5

-10

average of 10 hESCs

aver

age

of 4

9 hi

PS

Cs

C

D E

CTHRC1

ECRG4

PIGZ

DPYS

LPA

CD14

VAMP5

C6orf150

ACOT4

TERC

TERC

NPY

NPY

CXCL5

TACR3

TACR3

CALCB

MX1

NMBR

SLC6A5

MX1

HSPB6

IRAK4

IRAK4

CTHRC1

DPYS

ZNF354C

ACOT2

CFLAR

C17orf57

C6orf150

NMBR

MTNR1B

PON3PON3

VAMP5

HSPB6

ECRG4

KIAA1944KIAA1944FZD10

TCERG1LTCERG1L

SLC35E3

ZNF354C

C9orf64

CSRP1

TRIM4

CSRP1

SLITRK2

MOS

DHX34TAOK2

ZNF228

DHPSCOPZ1

ZNF228

NPHS2

GRM1

0 1

(DNA Methylation)

Gro

up A

Gro

up B

Gro

up C

COMT

DCDC2DCDC2

KAAG1CCDC68

C14orf62

LOC63928

hEC

Cs

canc

er c

ell l

ines

som

a

hESCs hiPSCs

Kyo

to U

niv.

othe

rs

Ret

rovi

rus

Epi

som

al

pla

smid

Sen

daiv

irus

0.5-0.5 0

ES

ave

rage

- iP

S a

vera

ge

F

G

hESCs hiPSCs

Met

hyla

tion

(%)

100

80

60

40

20

0

Met

hyla

tion

(%)

100

80

60

40

20

0

PON3 (Group A)

TCERG1L (Group C)

DNA methylation

average of 10 hESCs

aver

age

of 4

9 hi

PS

Cs

1.0

0.8

0.6

0.4

0.2

0 1.00.80.60.40.20

MNS1

TUSC1

PON3

LOC440905

KLHL34

RLN2

TRIM4LOC387647

PIP5K1C

AAK1

QRFP

lincRNA (chr.18)

lincRNA (chr.3)

lincRNA (chr.6)

lincRNA (chr.6)

C4orf49

LOC729970

TMBIM4

WDR1

CNDP2

ARL6IP6

COASY

SUPT5HAP3S1

SNX7

CYTH2

CXXC1

GCC2

ZNF335

PIGO

LOC100129250

RBM23

LOC100505876

DNAJC15

PXMP4

LRRC61

KLF8

lincRNA (chr.X)

lincRNA (chr.11)

AGBL4

A_33_P3232861

FAM171BTRIM6

SNX21

lincRNA (chr.X)

EXD3

lincRNA (chr.X)lincRNA (chr.13)

MAFIP

XLOC_004284

ANKRD7

LOC100130232

SDR42E1

OLIG2

ZNF441

FRMD3

LOC441177

RGPD1

TMEM132CKLF8

TRIM4

-10 -6 0 6-8 -4 -2 2 4

Abs

olut

e va

lue

of fo

ld c

hang

e(h

igh)

(low

)

mRNA expression

hiPSCs (49 clones)hESCs (10 clones)MAX

min avera

ge

Kyo

to U

niv.

othe

rs

Ret

rovi

rus

Epi

som

al p

lasm

id

Sen

daiv

irus

hESCs hiPSCs

Kyo

to U

niv.

othe

rs

Ret

rovi

rus

Epi

som

al p

lasm

id

Sen

daiv

irus

Fig. 1. hiPSCs and hESCs have overlapped variations in RNA expression and DNA methylation. Scatter plots of mRNA expression (A), miRNA expression (B),and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC lines (x axis). The RNA expression value is shown ona log 2 scale. Green lines indicate twofold differences in the RNA expression levels between the clones. Differentially expressed probes (t test, FDR < 0.05) areshown in magenta. (D) The variations in the mRNA expression levels of 61 differentially expressed probes in hESCs (red) and hiPSCs (black) are shown. Probesare arranged in order of the absolute value of the FC between hESCs and hiPSCs. (E) The DNA methylation profiles for CpGs contained in reported hES-hiPSDMRs and overlapping with our platform. Probes are arranged in order of the differences between the average DNA methylation level of hESCs and that ofhiPSCs. The heat map represents the DNA methylation levels from completely methylated (1, magenta) to unmethylated (0, white) samples. The methylationstatus of the upstream region of PON3 (F) and TCERG1L (G) was examined by pyrosequencing.

20570 | www.pnas.org/cgi/doi/10.1073/pnas.1319061110 Koyanagi-Aoi et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1

Page 3: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

distinguish some, but not all, hESCs from hiPSCs. SeventeenCpGs belonged to group B, which showed similar methylationlevels in both hESCs and hiPSCs. Five CpGs, representingthree genes, belonged to group C and showed higher meth-ylation levels in some, but not all, hiPSCs compared with hESCs.The remaining hiPSCs showed low methylation levels, comparableto those in hESCs. We confirmed the methylation status of a rep-resentative example of CpGs in group C, the upstream region ofthe transcription elongation regulator 1-like (TCERG1L), bypyrosequencing; the methylation levels were low in 21 of the 49hiPSCs (Fig. 1G). Therefore, the CpGs in group Cmay distinguishsome, but not all, iPSCs from hESCs.A previous report (12) showed that many CG-DMRs were

located in CpG shores, rather than CpG islands. Because onlya few (9 out of 71) of their CG-DMRs were covered by ourInfinium platform, we analyzed five CG-DMRs in CpG shores bypyrosequencing, including ataxin-2 binding protein 1 (A2BP1),insulin-like growth factor 1 receptor (IGF1R), POU domain,class 3, transcription factor 4 (POU3F4), protein tyrosine phos-phatase, receptor type, T (PTPRT), and zinc finger protein 184(ZNF184) (Fig. S1C). We detected significant differences in theaveraged DNA methylation levels between hESCs and hiPSCs infour out of five genes. However, variations of DNA methylationlevels in hESCs and hiPSCs are overlapped. They may distin-guish some, but not all, hiPSCs from hESCs.

A Subset of hiPSC Clones Retain Undifferentiated Cells After NeuralDifferentiation. To examine whether hESCs and hiPSCs havecomparable differentiation potential, we performed in vitro di-rected differentiation into neural stem and progenitor cells usingthe modified serum-free floating culture of embryoid body-likeaggregates (SFEBq) method (Fig. 2A) (18). We initially per-formed the neural induction of two hESCs and 21 hiPSCs.Fourteen days after induction, the differentiation efficiency wasevaluated based on the expression of an early neural marker,polysialylated neural cell adhesion molecule (PSA-NCAM). Wefound that all hESCs and hiPSCs differentiated into PSA-NCAM+ cells with more than 80% efficiency (Fig. 2B). We alsoquantified the expression levels of the early neural marker pairedbox 6 (PAX6) and the late neural marker microtubule-associatedprotein 2 (MAP2) in neurospheres by quantitative RT-PCR(qRT-PCR) (Fig. S2A). All of the examined hES/iPSCs expressedPAX6 at >100-fold higher levels and MAP2 at >20-fold higherlevels in comparison with undifferentiated H9 hESC. However, insome hiPSCs, we noticed slightly lower differentiation efficiencythan in the remaining hiPSCs and hESCs (Fig. 2B). This lowerefficiency in neural differentiation was inversely correlated witha higher proportion of POU class 5 homeobox 1 (POU5F1, alsoknown as OCT3/4)+ and TRA1-60+ undifferentiated cells (Fig.

2C). We also detected residual undifferentiated cells after a dif-ferent neural differentiation protocol using adhesion culture (19)(Fig. S2B).We then increased the number of clones and examined the

proportions of OCT3/4+ undifferentiated cells after neural in-duction from 10 hESCs and 40 hiPSCs. The 50 clones wereranked according to their proportions of OCT3/4+ cells on day14 (Dataset S1 and Fig. 2D). The proportions of OCT3/4+ cellsvaried from 0 to ∼20%. Thirty-eight clones, including ninehESCs and 29 hiPSCs, showed less than 1% OCT3/4+ cells inall experiments. We designated these clones as “good” clones.However, seven hiPSCs contained more than 10% OCT3/4+cells after neural differentiation in at least one experiment.We designated these clones as “differentiation-defective” clones.Clones that were not good or defective were categorized as“intermediate.”

Activation of Specific LTR7 Elements in Differentiation-DefectiveClones. To identify molecular signatures that can predict differ-entiation-defective clones, we compared the global gene expres-sion patterns of 38 good clones and seven differentiation-defectiveclones under the culture conditions used for the undifferentiatedstate. We identified 19 probes (13 putative transcripts) thatshowed greater than fivefold differences in expression, with anFDR <0.05, shown by magenta dots in Fig. 3A and listed inTable S1B.Of the 19 probes identified, five probes recognized HHLA1

(human endogenous retrovirus-H LTR-associating 1). Previousreports have shown that HHLA1 is regulated by a long terminalrepeat (LTR) of a human endogenous retrovirus-H (HERV-H)(20). The LTR inHHLA1 is classified as LTR7.Moreover, amongthe genes recognized by the 19 probes, we found that at least twoothers, abhydrolase domain containing 12B (ABHD12B) andchromosome 4 open reading frame 51 (C4orf51), also containedLTR7 sequences in their gene bodies. According to a microarrayanalysis, we confirmed that these three LTR7-containing geneswere up-regulated in the differentiation-defective hiPSCs, aswell as the nullipotent hECC line 2102Ep 4D3 (21), but theywere expressed at lower levels in the good hiPSCs, hESCs, andpluripotent hECC line NTera2 cloneD1. They were almost notexpressed in the original somatic cells (Fig. 3B).The Agilent Technologies microarray platform has 12 probes,

including two reverse probes [d(r), f(r)], for HHLA1 and itsneighboring gene, otoconin 90 (OC90), which is reported tomake a fusion transcript with HHLA1 (20) (Fig. 3C). Amongthem, seven probes located downstream of LTR7 showed higherexpression levels in differentiation-defective clones than in goodclones (Fig. 3 C and D). Similarly, there are two probes forABHD12B, designed for exons 4 and 13 (Fig. 3C). Only the exon

“defective” “good”

<SFEBq method>

SB431542+Dorsomorphin+Y27632

hES/iPSCs Flow cytometryA

B C

OC

T3/4

pos

itive

cel

ls (%

)

10

20

10

TKC

BV

5-6

(6)

TIG

107-

3f1

(4)

TIG

108-

4f3

(7)

451F

3 (7

)

TIG

118-

4f1

(6)

1375

-4f1

(2)

610B

1 (2

)60

4A1

(2)

TIG

120-

4f1

(2)

TIG

107-

4f1

(3)

TIG

114-

4f1

(2)

H9

(6)

1488

-4f1

(2)

246G

1 (2

)

703A

1 (3

)66

5A1

(3)

KhE

S1

(9)

TKC

B7-

2 (3

)20

1B2

(2)

TKC

BV

4-2

(3)

703B

1 (3

)

711A

6 (2

)71

1A1

(2)

606B

1 (2

)60

6A1

(2)

253G

4 (2

)

414C

2 (3

)

409B

2 (2

)

201B

7 (7

)

622G

1 (2

)60

4A3

(2)

711C

1 (2

)66

5A7

(3)

622E

1 (2

)61

0A2

(2)

604B

1 (2

)

585A

1 (3

)

454E

2 (3

)

404C

2 (2

)

253G

1 (3

)

1503

-4f1

(3)

201B

6 (2

)

KhE

S3

(1)

H1

(2)

KhE

S4

(3)

KhE

S5

(3)

ES

03 (3

)

ES

04 (3

)

ES

06 (3

)

SA

02 (2

)

PS

A-N

CA

M (+

) cel

ls (%

)

KhE

S1

H9

201B

220

1B7

253G

125

3G4

TIG

107-

4f1

TIG

107-

3f1

TIG

108-

4f3

TIG

114-

4f1

TIG

118-

4f1

TIG

120-

4f1

1375

-4f1

1488

-4f1

246G

1TK

CB

V4-

2TK

CB

V5-

6TK

CB

7-2

404C

240

9B2

414C

245

1F3

454E

2

Exp.1 Exp.2

hESCs hiPSCs

HDF HDFCB DP(origin)(method) Retrovirus Episomal

10080604020

0

10080604020

0 pos

itive

cel

ls (%

)

H9

201B

2

TIG

108-

4f3

TIG

118-

4f1

TKC

BV

5-6

hESC hiPSCs

D

PSA-NCAMOCT3/4 TRA1-60

Fig. 2. A differentiation-defective phenotype in a subset of hiPSC clones. (A) A schematic diagram of the SFEBq method used for neural differentiation. (B)Neural induction was performed for 2 hESC and 21 hiPSC lines which were established from various origins by retroviral or episomal vector methods. On day14, we examined the proportion of PSA-NCAM–expressing cells by flow cytometry (n = 2). (C) The proportions of PSA-NCAM- (white), OCT3/4- (gray), andTRA1-60-positive (black) cells 14 d after neural differentiation. (D) The proportions of OCT3/4-positive cells on day 14 after neural differentiation are ranked inorder of their maximum value. The numbers in parentheses show the number of trials.

Koyanagi-Aoi et al. PNAS | December 17, 2013 | vol. 110 | no. 51 | 20571

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1

Page 4: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

13 probe, located downstream of LTR7, showed a higher ex-pression in differentiation-defective clones than in good clones(Fig. 3D). We also performed an exon array (Affymetrix) ofABHD12B and C4orf51 and found that exons downstreamof LTR7 were preferentially up-regulated in differentiation-defective hiPSC clones (Fig. 3E). We also found that themethylation status of LTR7s in these three genes were lower indifferentiation-defective hiPSC clones than in good clones (Fig.3F). These results indicate that the three genes are transcribedfrom activated LTR7.

DNA Hypomethylation Exists in Some, but Not All, LTR7s inDifferentiation-Defective Clones. According to the Repeatmaskersoftware program, there are 3,523 LTR7 elements in the humangenome. To extract microarray probes that are potentially af-fected by LTR7s, we first selected genes containing LTR7s intheir gene bodies or regions 2 kb upstream from their tran-scription start sites. We then retrieved the microarray probeslocated between each LTR7 and the 3′ end of the correspondinggene body. As a result, we selected 763 probes as LTR7-relatedprobes (Fig. S3A and Table S1C). We found that most of theseprobes showed comparable expression levels in good and de-fective lines (Fig. S3B), with the exception of some probes, suchas those corresponding to arrestin, beta 1 (ARRB1), fatty acidamide hydrolase 2 (FAAH2), and TBC1 domain family, member23 (TBC1D23), that were differentially expressed between goodand defective clones (FDR <0.05 and FC >2) and showedslightly higher expression in defective lines.We then checked the DNA methylation status of the LTR7

regions in these three genes and three other genes DNA (cyto-sine-5-)-methyltransferase 3 beta (DNMT3B), ATP-binding cas-sette, sub-family A (ABC1), member 1 (ABCA1), and amyloidbeta (A4) precursor protein (APP)] whose expression levels werenot significantly different between the good and defective clones.By pyrosequencing and clonal bisulfite sequencing, we found thatthe LTR7 regions in four genes (ARRB1, FAAH2, TBC1D23,and APP) were hypomethylated in defective clones comparedwith good clones. In contrast, the LTR7 regions in two genes

(DNMT3B and ABCA1) did not show such hypomethylation (Fig.S3C). Therefore, the activation of LTR7 is not confined toHHLA1, ABHD12B, and C4orf51; DNA hypomethylation existsin some, but not all, LTR7s in defective hiPSCs.

Differentiation-Defective hiPSC Clones Form Teratomas in MouseBrains. To further evaluate the defective hiPSCs, we inducedtheir differentiation into dopaminergic neurons, which were thentransplanted into the striata of nonobese diabetic/severe com-bined immune-deficient (NOD/SCID) mouse brains (Fig. 4A).Thirty and 60 d after transplantation, we obtained T2-weightedimages of the mouse brains with an MRI scanner to observe thegraft sizes at the transplanted sites (Fig. 4B). The quantificationof the MRI images showed that defective hiPSC clones resultedin significantly larger graft sizes than good clones (Fig. 4C).Notably, some mice that had received defective clones died ordeveloped symptoms that required euthanasia before day 60(Table S2). Therefore, we could not obtain the graft size data onday 60 in these mice.To identify the composition of the surviving grafts, we per-

formed a histological analysis of the brains of animals that diedor that became moribund after transplantation. The remaininghealthy mice were euthanized 14–41 wk after transplantation.Sections were stained with H&E. Thirty-six of the 42 grafts(85.7%) from defective clones contained nonneural lineage tis-sues, such as intestine-like epithelial cells, cartilage, or mes-enchymal cells (Fig. 4D and Fig. S4A). In contrast, grafts fromgood clones largely consisted of neural tissues. Immunostainingconfirmed grafts were positive for human neural cell adhesionmolecule (NCAM) (Fig. S4B). A qRT-PCR analysis of pre-transplanted cells from defective clones revealed higher expres-sion levels of OCT3/4, suggesting that some undifferentiatedcells still remained even after 29 d of neural induction (Fig. 4E).We then depleted the TRA-1-60+ cells on day 22 during neuralinduction and transplanted cells on day 29. The TRA-1-60–depletedcells from defective clones resulted in significantly smaller graftsthat did not contain nonneural tissues (Fig. S4C).

B

420-2-4-6

420

-2-4-6

DC

“defective” (n=3)

E

F

mRNA expressionAFDR<0.05, FC >5

19 probes

HHLA1

ABHD12BC4orf51

(2 CpGs)5’ LTR

(4 CpGs)3’ LTR

(2 CpGs)

5’ LTR(2 CpGs)

3’ LTR(3 CpGs)

Met

hyla

tion

(%)

HHLA1 ABHD12B C4orf51

↓ ↓

EFR3A

KCNQ3

abc

d(r) ef(r) g

hij

k

l n

o

“defec

tive”

(n=6)

“good

” (n=

6)

NTera2

2102

EpHDF DP

3’ LTR

“defec

tive”

(n=6)

“good

” (n=

6)

NTera2

2102

EpHDF DP

“defec

tive”

(n=6)

“good

” (n=

6)

NTera2

2102

EpHDF DP

C4orf51

HHLA1 & OC90

ABHD12B

HERV-H intLTR7 Exon

egh

ijk l

Microarray probe

n

o

ABHD12B

“defective” “intermediate” “good”hECCs soma cancer cell lines

“good”(n=3)

soma(n=2)

86420-2-4-6

86420

-2-4-6

6

0

-6

6

0

-6

0

-6

Nor

mal

ized

inte

nsity

(log

2)

HHLA1

ABHD12B

C4orf51

Exp

ress

ion

leve

l (lo

g2) C4orf51

“defective” (n=7)

“goo

d” (

n=38

)

100

0

50

100

0

50

100

0

50

***** *

141210

86420 ▲

LTR71210

86420 ▲

LTR7

n.s.

m

1 2 3 4 5 6exon

2 3 4 6 7 8 9 10 11 12 13exon

2102Ep

NTera2

“defective” (n=7)

a bc d(r)f(r)

m

1 2 3 45 6

1 23 4 5 678 9 10 111213 “goo

d” (

n=38

)

hEC soma hEC soma hEC soma

Fig. 3. Activation of specific endogenous retroviral LTR7s in defective clones. (A) A scatter plot of the mRNA expression data comparing the average of 38good clones (y axis) to the average of seven defective clones (x axis). Green lines indicate fivefold differences in expression. A total of 19 differentiallyexpressed probes are colored magenta. (B) The expression levels of LTR7-related genes (HHLA1, ABHD12B, and C4orf51) were examined by microarray. (C) Aschematic diagram of three LTR7-related genes. HHLA1 and OC90 are neighboring genes. Dots indicate microarray probes. Magenta dots show probes thatare located after LTR7 regions, which were up-regulated in defective clones. (D) A scatter plot of array probes that recognized LTR7-related genes and twoother genes [EFR3 homolog A (S. cerevisiae, EFR3A) and potassium voltage-gated channel, KQT-like subfamily, member 3 (KCNQ3)], which are genesneighboring HHLA1 and OC90, respectively. (E) The exon array of the ABHD12B and C4orf51. The average levels of the normalized exon expression areshown. (F) The DNA methylation status of LTR7 and its neighboring regions of HHLA1, ABHD12B, and C4orf51 was examined by pyrosequencing. (n.s., notsignificant; *P < 0.05, **P < 0.01, Mann–Whitney U test).

20572 | www.pnas.org/cgi/doi/10.1073/pnas.1319061110 Koyanagi-Aoi et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1

Page 5: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

We also observed that 14 out of the 63 (22.2%) grafts fromgood clones, including those from hESCs, contained a nonneuralcomponent in the graft tissue after transplantation (Fig. 4D andTable S2), although these clones did not show high expressionlevels of OCT3/4 in the pretransplantation samples (Fig. 4E). Wereferred to these clones as “type-2 defective” clones, which weredistinct from “type-1 defective” clones that contained OCT3/4+

undifferentiated cells in the pretransplantation samples. Weobserved higher expression levels of SRY (sex determiningregion Y)-box 17 (SOX17, an endoderm marker) and goose-coid homeobox (GSC, an endoderm and mesoderm marker) inthe pretransplantation samples of type-2 defective clones (Fig. 4F and G), demonstrating the presence of other lineages in thesepretransplantation samples.

DiscussionWe identified two types of defective pluripotent stem cell linesin this study. The first type consisted of hiPSCs that retaineda substantial number of undifferentiated cells after in vitro di-rected neural differentiation. Seven out of the 40 iPSCs (17.5%)examined in this study fell into this category. In contrast, we didnot observe such defects in any of the 10 hESCs. More clonesshould be analyzed to confirm that hESCs are free from thisdeficiency. Nevertheless, it is likely that type-1 defectiveness ismore common in hiPSCs than in hESCs. The type-1 defectivehiPSCs are accompanied by an aberrant epigenetic status. Amongthe 13 putative transcripts that were highly expressed in thesedefective clones, at least three were expressed from the LTR ofendogenous retroviruses. Normally, these LTRs are silenced byvarious epigenetic modifications, including DNA methylation (22–24). In type-1 defective iPSC clones, the LTR locus in the threegenes showed lower DNA methylation levels than in good clonesand original somatic cells. Notably, the same regions were hypo-methylated in the nullipotent hECC line, 2102Ep 4D3, suggestingthat the loss of DNA methylation in these LTR loci is correlatedwith the lower ability to differentiate. At present, the biologicalsignificance and relationship between activation of specific LTRsand the defective phenotype is unclear. Recent reports showedthat endogenous retrovirus may play roles in the establishmentand maintenance of transcription network in pluripotent stemcells (25, 26). Furthermore, updated annotations revealed that

one of the differentially expressed probes in type-1 defectivehiPSCs (A_19_P00325604) encoded large intergenic noncodingRNA regulator of reprogramming (Linc-ROR), which containedLTR7 in its 5′ region. Linc-ROR is reported to have multipleroles in the induction and maintenance of pluripotency (27, 28).Future studies should be undertaken to clarify why these epi-genetic abnormalities occur and how they are related to thedefective differentiation.Kim et al. (29) showed that there is an inverse correlation

between the hsa-mir-371–373 expression and the efficiency ofneural differentiation. They also showed that Kruppel-like factor4 (gut) (KLF4) may induce the expression of hsa-mir-371–373. Inour analyses, the hsa-mir-371–373 cluster was highly expressed inall of the seven type-1 defective hiPSC clones (Fig. S5A). How-ever, the cluster was also highly expressed in many good clones.KLF4was highly expressed in some defective clones (Fig. S5B), andfour out of six retroviral defective clones failed to silence KLF4retroviral transgenes (Fig. S5C). There was no correlation betweenthe OCT3/4 transgene expression and type-1 defectiveness (Fig.S5D). Taken together, these findings indicate that high expres-sion levels of the hsa-mir-371–373 cluster, KLF4, and transgenescannot function as absolute markers for type-1 defectiveness.We previously reported that the origin of mouse iPSCs was

a major determinant of defectiveness in directed neural differ-entiation; mouse iPSCs from adult tail tip fibroblasts showed thehighest incidence of resistance to differentiation (30). In thepresent study using human iPSCs, five out of seven type-1 de-fective clones were derived from fibroblasts of donors of variousages, and six out of the seven clones were generated using ret-roviruses (Table 1). This may suggest that type-1 defectiveness isassociated with fibroblast origin and retroviral induction. How-ever, in this study, most of the fibroblast-derived iPSCs weregenerated by retroviruses, and most of the nonfibroblast iPSCswere generated by nonretroviral methods. Future studies willneed to be undertaken to determine whether the origin or thegeneration method (or both) has a significant impact on thefrequency of type-1 differentiation-defective iPSCs.The second type of defective group includes hiPSCs and

hESCs that contained differentiated cells of nonneural lineagesafter in vitro directed differentiation into dopaminergic neurons.We have previously shown that the optimal conditions for hepatic

DC

B

A

Day 59

Day 89

Gra

ft se

ctio

n su

rface

(mm

)2

*

*40

30

20

10

0

“defec

tive”

“inter

mediat

e”

“good

“defec

tive”

“inter

mediat

e”

“good

“defec

tive”

“inter

mediat

e”

“good

”Neu

ral t

issu

e pr

opor

tion

(%)

201B7TIG107-3f1 451F3“defective” “good”

OCT3/4

F

G

SOX17

TKC

BV

5-6

TIG

118-

4f1

TIG

108-

4f3

1503

-4f1

TIG

107-

3f1

451F

3TI

G10

7-4f

160

4A1

610B

1K

hES1

604B

1

TKC

BV

4-2

TKC

B7-

2H

9

606A

160

6B1

253G

420

1B7

KhE

S360

4A3

409B

2

H1

585A

1

414C

225

3G1

610A

2

454E

2

201B

6

som

a

GSC

“type2defective”

“type1defective”

ES

C

“defective” “good”

42

68

101214

0

mR

NA

exp

ress

ion

E

ES/iPSC

SB431542Dorsomorphin

Y27632

Growthfactors

Day 59/89MRI Scan

(x10 )6

0

5

10

15

20

25

42

68

1012

0

(x10 )2

(x10 )2“type2

defective”“type1

defective”

mR

NA

exp

ress

ion

m

RN

A e

xpre

ssio

n

Day 29Transplantation

<40%<60%<80%<99%100%unknown

0

50

100

Fig. 4. Transplantation of neural cells derived fromhiPSCs and hESCs into mouse brains. (A) A schematicdiagram of the SFEBq method used for dopami-nergic neural differentiation. On day 29, the cellswere transplanted into NOD/SCID mouse brains. (B)Magnetic resonance images of coronal sections ofthe grafted brains. The section surface of graftedcells indicated as the white shadow in right brainwas measured as described in the lower panels. (C)A box-and-whisker plot of the surface sizes of graftsections 30 and 60 d after transplantation. Themedian, quartile, and range are shown. *P < 0.05(t test). (D) The proportion of each kind of graft.Grafts were categorized according to their compo-nents as determined in H&E sections and wereclassified by the proportion of neural tissues bya microscopic observation. The expression levels ofthe undifferentiated cell marker, OCT3/4 (E), theendoderm marker, SOX17 (F), and the mesodermand endodermmarker, GSC (G), in pretransplantationcultures, undifferentiated hESC lines, and somaticcells (HDF and DP) were examined by qRT-PCR. Thecolors of the dots were identical to the proportionof neural tissues in D.

Koyanagi-Aoi et al. PNAS | December 17, 2013 | vol. 110 | no. 51 | 20573

CELL

BIOLO

GY

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1

Page 6: Differentiation-defective phenotypes revealed by large ...Oct 30, 2013  · and DNA methylation (C) data comparing the average of 49 hiPSC lines (y axis) to the average of 10 hESC

differentiation are different for each clone (16). By optimizingthe protocols, it may be possible to induce complete neural dif-ferentiation to avoid type-2 defective clones. Alternatively, puri-fication of neural cells using a cell sorter may work to avert type-2defective clones.Several studies have reported sets of genes whose DNA

methylation status is different between hiPSCs and hESCs. Wevalidated these CG-DMRs and found that many of them candistinguish some hESCs from hiPSCs (group A in Fig. 1E). Theyare highly methylated in some hESCs, but not in hiPSCs ororiginal somatic cells. Thus, these CG-DMRs may representepigenetic memories of somatic cells in iPSCs. However, wefound a set of hESCs that showed low methylation status of theseCG-DMRs, which were comparable to hiPSCs. We also foundanother set of the reported CG-DMRs that showed high meth-ylation status in some hiPSCs but not in original somatic cells orhESCs (group C in Fig. 1E). These likely represent aberrantmethylation associated with reprogramming. However, we alsofound many hiPSCs showed normal methylation patterns ofthese CG-DMRs. A more recent study identified nine genesthat can segregate hiPSCs from hESCs in DNA methylationand gene expression (31). However, we did not observe such aclear distinction in gene expression of these genes betweenour hiPSCs and hESCs (Fig. S6). Two of these genes, TCERG1Land FAM19A5, may distinguish some, but not all, hiPSCsfrom hESCs.In our analyses, 35 hiPSCs had records of the donor’s genetic

background; 14 were derived from Caucasians and 21 were fromJapanese subjects (Dataset S1). Thus, the similarity of somesignatures between the Kyoto hESCs and our hiPSCs cannot beattributed to the racial or ethnic backgrounds of the donors.Another possible cause of the differences is the method used toestablish the hESCs and the subsequent culture conditions. TheKyoto hESCs were generated on feeders consisting of a 1:1mixture of mouse embryonic fibroblasts and SL10 cells (17, 32),which were subcloned from STO cells. Most of our hiPSCs were

established on SNL feeders, which were also derived from STOcells. A recent report showed that the feeders have profoundeffects on established hiPSCs (33). To confirm the importance ofthe culture conditions, more studies comparing hESC/hiPSCsestablished under different conditions will be needed.In conclusion, our results revealed that a subset of hiPSCs is

defective in neural differentiation and marked with activation ofendogenous retroviruses. We also confirmed that some hiPSCsare different from hESCs in molecular signatures, including CG-DMRs, which has been previously reported. It remains to bedetermined whether these molecular signatures specific for somehiPSCs have functional consequences.

Materials and MethodsGene expression profiling was carried out using the SurePrint G3 human GEmicroarray (Agilent Technologies). Most of the data were analyzed using theGeneSpring GX 11.5.1 software program (Agilent Technologies). Neural in-duction was performed as described previously (18). Detailed descriptions ofmethods are available in SI Materials and Methods.

ACKNOWLEDGMENTS. We thank Drs. Takashi Aoi, Yoshinori Yoshida, MasatoNakagawa, and other members of the S.Y. research group for their valuablescientific discussions; Drs. Takuya Yamamoto, Toshiki Taya, and TomoakiTsubota for advice about bioinformatics; Dr. Hirofumi Suemori for hES celllines; Drs. Koji Eto and Naoya Takayama for TKCB hiPS cell lines; AkikoOtsuka for RNA and genomic DNA samples; Dr. Takafumi Kimura andTakaomi Futakami for HLA analysis; and Aki Sasaki, Tomomi Ito, MidoriYokura, and Ayumi Ichikawa for technical assistance. We also thank NaokiNagata, Yoko Miyake, Rie Kato, Eri Minamitani, Sayaka Takeshima, andRyoko Fujiwara for their valuable administrative support. This work wassupported in part by Grants-in-Aid for Scientific Research from the JapaneseSociety for the Promotion of Science; from the Ministry of Education,Culture, Sports, Science, and Technology; by a grant from the LeadingProject of the Ministry of Education, Culture, Sports, Science, and Technol-ogy; and by a grant from the Funding Program for World-Leading InnovativeResearch and Development on Science and Technology (First Program) of theJapanese Society for the Promotion of Science. M.K.-A. and M.O. wereResearch Fellows of the Japanese Society for the Promotion of Science.

1. Thomson JA, et al. (1998) Embryonic stem cell lines derived from human blastocysts.Science 282(5391):1145–1147.

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

3. Yu J, et al. (2007) Induced pluripotent stem cell lines derived from human somaticcells. Science 318(5858):1917–1920.

4. Bock C, et al. (2011) Reference maps of human ES and iPS cell variation enable high-throughput characterization of pluripotent cell lines. Cell 144(3):439–452.

5. Newman AM, Cooper JB (2010) Lab-specific gene expression signatures in pluripotentstem cells. Cell Stem Cell 7(2):258–262.

6. Guenther MG, et al. (2010) Chromatin structure and gene expression programs ofhuman embryonic and induced pluripotent stem cells. Cell Stem Cell 7(2):249–257.

7. Chin MH, et al. (2009) Induced pluripotent stem cells and embryonic stem cells aredistinguished by gene expression signatures. Cell Stem Cell 5(1):111–123.

8. Marchetto MC, et al. (2009) Transcriptional signature and memory retention of hu-man-induced pluripotent stem cells. PLoS ONE 4(9):e7076.

9. Ghosh Z, et al. (2010) Persistent donor cell gene expression among human inducedpluripotent stem cells contributes to differences with human embryonic stem cells.PLoS ONE 5(2):e8975.

10. Ohi Y, et al. (2011) Incomplete DNA methylation underlies a transcriptional memoryof somatic cells in human iPS cells. Nat Cell Biol 13(5):541–549.

11. Deng J, et al. (2009) Targeted bisulfite sequencing reveals changes in DNA methyla-tion associated with nuclear reprogramming. Nat Biotechnol 27(4):353–360.

12. Doi A, et al. (2009) Differential methylation of tissue- and cancer-specific CpG islandshores distinguishes human induced pluripotent stem cells, embryonic stem cells andfibroblasts. Nat Genet 41(12):1350–1353.

13. Lister R, et al. (2011) Hotspots of aberrant epigenomic reprogramming in humaninduced pluripotent stem cells. Nature 471(7336):68–73.

14. Hu BY, et al. (2010) Neural differentiation of human induced pluripotent stem cellsfollows developmental principles but with variable potency. Proc Natl Acad Sci USA107(9):4335–4340.

15. Yamanaka S (2012) Induced pluripotent stem cells: Past, present, and future. CellStem Cell 10(6):678–684.

16. Kajiwara M, et al. (2012) Donor-dependent variations in hepatic differentiation fromhuman-induced pluripotent stem cells. Proc Natl Acad Sci USA 109(31):12538–12543.

17. Suemori H, et al. (2006) Efficient establishment of human embryonic stem cell linesand long-term maintenance with stable karyotype by enzymatic bulk passage. Bio-chem Biophys Res Commun 345(3):926–932.

18. Morizane A, Doi D, Kikuchi T, Nishimura K, Takahashi J (2011) Small-molecule in-hibitors of bone morphogenic protein and activin/nodal signals promote highly ef-ficient neural induction from human pluripotent stem cells. J Neurosci Res 89(2):117–126.

19. Kriks S, et al. (2011) Dopamine neurons derived from human ES cells efficiently en-graft in animal models of Parkinson’s disease. Nature 480(7378):547–551.

20. Kowalski PE, Freeman JD, Mager DL (1999) Intergenic splicing between a HERV-Hendogenous retrovirus and two adjacent human genes. Genomics 57(3):371–379.

21. Bahrami AR, Matin MM, Andrews PW (2005) The CDK inhibitor p27 enhances neuraldifferentiation in pluripotent NTERA2 human EC cells, but does not permit differ-entiation of 2102Ep nullipotent human EC cells. Mech Dev 122(9):1034–1042.

22. Rowe HM, Trono D (2011) Dynamic control of endogenous retroviruses during de-velopment. Virology 411(2):273–287.

23. Hutnick LK, Huang X, Loo TC, Ma Z, Fan G (2010) Repression of retrotransposal ele-ments in mouse embryonic stem cells is primarily mediated by a DNA methylation-independent mechanism. J Biol Chem 285(27):21082–21091.

24. Stoye JP (2012) Studies of endogenous retroviruses reveal a continuing evolutionarysaga. Nat Rev Microbiol 10(6):395–406.

25. Macfarlan TS, et al. (2012) Embryonic stem cell potency fluctuates with endogenousretrovirus activity. Nature 487(7405):57–63.

26. Santoni FA, Guerra J, Luban J (2012) HERV-H RNA is abundant in human embryonicstem cells and a precise marker for pluripotency. Retrovirology 9:111.

27. Loewer S, et al. (2010) Large intergenic non-coding RNA-RoR modulates reprogrammingof human induced pluripotent stem cells. Nat Genet 42(12):1113–1117.

28. Wang Y, et al. (2013) Endogenous miRNA sponge lincRNA-RoR regulates Oct4,Nanog, and Sox2 in human embryonic stem cell self-renewal. Dev Cell 25(1):69–80.

29. Kim H, et al. (2011) miR-371-3 expression predicts neural differentiation propensity inhuman pluripotent stem cells. Cell Stem Cell 8(6):695–706.

30. Miura K, et al. (2009) Variation in the safety of induced pluripotent stem cell lines. NatBiotechnol 27(8):743–745.

31. Ruiz S, et al. (2012) Identification of a specific reprogramming-associated epigeneticsignature in human induced pluripotent stem cells. Proc Natl Acad Sci USA 109(40):16196–16201.

32. Kawase E, et al. (1994) Strain difference in establishment of mouse embryonic stem(ES) cell lines. Int J Dev Biol 38(2):385–390.

33. Tomoda K, et al. (2012) Derivation conditions impact X-inactivation status in femalehuman induced pluripotent stem cells. Cell Stem Cell 11(1):91–99.

20574 | www.pnas.org/cgi/doi/10.1073/pnas.1319061110 Koyanagi-Aoi et al.

Dow

nloa

ded

by g

uest

on

Feb

ruar

y 9,

202

1