+ All Categories
Home > Documents > Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in...

Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in...

Date post: 22-Jan-2021
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
13
REGULAR ARTICLE Interspecies chimera between primate embryonic stem cells and mouse embryos: Monkey ESCs engraft into mouse embryos, but not post-implantation fetuses Calvin Simerly a , Dave McFarland a , Carlos Castro a , Chih-Cheng Lin a , Carrie Redinger a , Ethan Jacoby a , 1 , Jocelyn Mich-Basso a , 2 , Kyle Orwig a , Parker Mills b , Eric Ahrens b , Chris Navara a , 3 , Gerald Schatten a , a Division of Developmental and Regenerative Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Pittsburgh School of Medicine, Pittsburgh Development Center, Magee-Womens Research Institute and Foundation, 204 Craft Avenue, Pittsburgh, PA, USA b Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA Received 11 August 2010; received in revised form 4 March 2011; accepted 10 March 2011 Available online 25 March 2011 Abstract Unequivocal evidence for pluripotency in which embryonic stem cells contribute to chimeric offspring has yet to be demonstrated in human or nonhuman primates (NHPs). Here, rhesus and baboons ESCs were investigated in interspecific mouse chimera generated by aggregation or blastocyst injection. Aggregation chimera produced mouse blastocysts with GFP-nhpESCs at the inner cell mass (ICM), and embryo transfers (ETs) generated dimly-fluorescencing abnormal fetuses. Direct injection of GFP-nhpESCs into blastocysts produced normal non-GFP-fluorescencing fetuses. Injected chimera showed N 70% loss of GFP-nhpESCs after 21 h culture. Outgrowths of all chimeric blastocysts established distinct but separate mouse- and NHP-ESC colonies. Extensive endogenous autofluorescence compromised anti-GFP detection and PCR analysis did not detect nhpESCs in fetuses. NhpESCs localize to the ICM in chimera and generate pregnancies. Because primate ESCs do not engraft post-implantation, and also because endogenous autofluorescence results in misleading positive signals, interspecific chimera assays for pluripotency with primate stem cells is unreliable with the currently available ESCs. Testing primate ESCs reprogrammed into even more naïve states in these inter-specific chimera assays will be an important future endeavor. © 2011 Elsevier B.V. All rights reserved. Abbreviations: PSC, pluripotent stem cells; ESC, embryonic stem cells; iPSC, induced pluripotent stem cells; nhpESC, nonhuman primate embryonic stem cells; ICM, inner cell mass cells. Corresponding author. Fax: + 1 412 641 2410. E-mail address: [email protected] (G. Schatten). 1 Present address: Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA. 2 Present address: Stem Cell Core, Rangos Children's Research, University of Pittsburgh 15213, USA. 3 Present address: Department of Biological Sciences, University of Texas, San Antonio, TX. 78249, USA. 1873-5061/$ see front matter © 2011 Elsevier B.V. All rights reserved. doi:10.1016/j.scr.2011.03.002 available at www.sciencedirect.com www.elsevier.com/locate/scr Stem Cell Research (2011) 7, 2840
Transcript
Page 1: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

ava i l ab l e a t www.sc i enced i r ec t . com

www.e l sev i e r . com/ l oca te / sc r

Stem Cell Research (2011) 7, 28–40

REGULAR ARTICLE

Interspecies chimera between primate embryonicstem cells and mouse embryos: Monkey ESCsengraft into mouse embryos, but notpost-implantation fetusesCalvin Simerly a, Dave McFarlanda, Carlos Castroa, Chih-Cheng Lina,Carrie Redinger a, Ethan Jacobya,1, Jocelyn Mich-Bassoa,2, Kyle Orwig a,Parker Mills b, Eric Ahrensb, Chris Navaraa,3, Gerald Schattena,⁎

a Division of Developmental and Regenerative Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences,University of Pittsburgh School of Medicine, Pittsburgh Development Center, Magee-Womens Research Institute and Foundation,204 Craft Avenue, Pittsburgh, PA, USAb Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA

Received 11 August 2010; received in revised form 4 March 2011; accepted 10 March 2011Available online 25 March 2011

Abstract Unequivocal evidence for pluripotency in which embryonic stem cells contribute to chimeric offspring has yet to bedemonstrated in human or nonhuman primates (NHPs). Here, rhesus and baboons ESCs were investigated in interspecific mousechimera generated by aggregation or blastocyst injection. Aggregation chimera produced mouse blastocysts with GFP-nhpESCsat the inner cell mass (ICM), and embryo transfers (ETs) generated dimly-fluorescencing abnormal fetuses. Direct injectionof GFP-nhpESCs into blastocysts produced normal non-GFP-fluorescencing fetuses. Injected chimera showed N70% loss ofGFP-nhpESCs after 21 h culture. Outgrowths of all chimeric blastocysts established distinct but separate mouse- and NHP-ESCcolonies. Extensive endogenous autofluorescence compromised anti-GFP detection and PCR analysis did not detect nhpESCsin fetuses. NhpESCs localize to the ICM in chimera and generate pregnancies. Because primate ESCs do not engraftpost-implantation, and also because endogenous autofluorescence results in misleading positive signals, interspecific chimeraassays for pluripotency with primate stem cells is unreliable with the currently available ESCs. Testing primate ESCsreprogrammed into even more naïve states in these inter-specific chimera assays will be an important future endeavor.

© 2011 Elsevier B.V. All rights reserved.

Abbreviations: PSC, pluripotent stem cells; ESC, embryonic stem celembryonic stem cells; ICM, inner cell mass cells.⁎ Corresponding author. Fax: +1 412 641 2410.E-mail address: [email protected] (G. Schatten).

1 Present address: Department of Obstetrics and Gynecology, DivisionHealth Science Center at San Antonio, San Antonio, TX 78229, USA.2 Present address: Stem Cell Core, Rangos Children's Research, Univers3 Present address: Department of Biological Sciences, University of Te

1873-5061/$ – see front matter © 2011 Elsevier B.V. All rights reserveddoi:10.1016/j.scr.2011.03.002

ls; iPSC, induced pluripotent stem cells; nhpESC, nonhuman primate

of Reproductive Endocrinology and Infertility, University of Texas

ity of Pittsburgh 15213, USA.xas, San Antonio, TX. 78249, USA.

.

Page 2: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

29Interspecies chimera between primate embryonic stem cells and mouse embryos

Introduction

Pluripotency is now recognized as a spectrum of biologicalplasticity rather than an ‘on–off’ toggle switch, and criteriafor assaying pluripotency range from the most demandingthrough to less stringent criteria. Certainly, the goldstandard assay involves chimera in which pluripotent stemcells, both embryonic stem cells (ESCs); (Lallemand andBrulet, 1990; Nagy et al., 1990; Wood et al., 1993) and morerecently PSCs (pluripotent stem cells); (Takahashi et al.,2006; Okita et al., 2007; Wernig et al., 2007) havecontributed to both offspring and germ cells after transferof either normally fertilized embryos or embryos generatedusing tetraploid complementation (Nagy et al., 1990; Eakinand Behringer, 2003). ESCs are colonies of self-renewingpluripotent cells that demonstrate the ability to differenti-ate into all three germ layers in the adult body (Evans andKaufman, 1981; Martin, 1981; Thomson et al., 1998). Theseand other PSCs promise therapeutic applications for humandisorders and diseases, and contribute further scientificallyas research resources for discovering the fundamentalmechanisms of human development and differentiation(reviewed by Riazi et al., 2009). Notwithstanding theirpotential medical importance, ethical constraints prohibitvital experiments to determine the safety, efficacy andtherapeutic potentials of human embryonic stem cells(hESCs); (Daley et al., 2007). Compelling arguments forprohibiting the use of human induced pluripotent stem cells(hiPSCs) in reproductive cloning in chimera have beenpublished (Lo et al., 2010), as have thoughtful considerationsof the biological merits and ethical constraints regardingusing human: animal chimera for biomedical research (Hyunet al., 2007; Behringer, 2007; Lensch et al., 2007).Consequently, there exist strong rationales for determiningthe full extent of pluripotentiality, as well as the biologicallimitations of human- and non-human-primate cells referredto as ‘pluripotent.'

Clinical extrapolations in stem cell medicine rest on thesolid scientific foundations of a quarter-century of investi-gations using mouse embryonic stem cells (mESCs) (reviewedby Evans, 2005). Yet several major concerns remain thatcannot be readily answered by studying hESC cell lines invitro or transplanted into relatively short-lived immunocom-promised mice. These questions include whether nhpESCshave full pluripotency as assayed in nonhuman primate (NHP)chimeras, whether differentiated cells remain committedafter transplantation and whether ESCs can proliferate ormigrate uncontrollably. Recently, important findings havebeen reported regarding PSC differentiation (Boyd et al.,2008; Trounson, 2006; Vaca et al., 2006; Mizuseki et al.,2003; Elkabetz et al., 2008; Kawasaki et al., 2002; Nakatsujiet al., 2008; Stadtfeld et al., 2008), therapeutic improve-ments after transplantation (Takahashi, 2006; Takagi et al.,2005); histocompatibility assays (Dighe et al., 2008; Rajeshet al., 2007); and epigenetics (Rugg-Gunn et al., 2005a,2005b; Zhang et al., 2007; Rugg-Gunn et al., 2007; Fujimotoet al., 2005, 2006; Mitalipov et al., 2007; Mitalipov, 2006).Lastly, the breakthrough discoveries of inducing pluripo-tency (iPS); (reviewed by Yamanaka, 2008) using human,nonhuman primate (Liu et al., 2008), and mouse cells havedramatically elevated the importance of pluripotency assaysfor both fundamental developmental biology as well as

medicine. Importantly, iPSCs from mice have been demon-strated to result in germ line transmission in both chimericembryo assays (Okita et al., 2007, 2008; Wernig et al., 2008)as well as in tetraploid complementation experiments(Meissner et al., 2007).

ESC pluripotency has most convincingly been demonstrat-ed in reaggregated embryos where the resultant offspringhave ESC contributions to all germ layers and tissues,including the germ line (reviewed by Rossant, 2001). Thusfar, only mouse and rat embryonic stem cells (mESCs)aggregated with mouse or rat embryos result in offspringborn with demonstrated ESC contribution to all three germlayers and the germ line (Iannaccone and Jacob, 2009).Currently, hESC differentiation is assayed by embryoidbodies (EBs) or teratomas and both contribute to all threegerm layers (Conley et al., 2005), but these technologieshave limitations—EBs do not mimic 3D axial morphogenesis invitro accurately and teratomas are a foreign environmentthat do not produce germ cells. Overwhelming ethicalconcerns obviously preclude interspecific chimera attemptswith hESCs. However, the derivation of nonhuman primateESCs (nhpESCs) can responsibly bridge gaps in our scientificknowledge between mESCs and hESCs, for example, in thegeneration of chimeric nonhuman primates with nhpESCscells, although issues with NHP embryo availability, cost, andcomplex technical obstacles with chimera production remain(Takada et al., 2002; Schramm and Paprocki, 2004; Scott,2006; Roberts, 2005). Thus, Mitalipov et al. (Mitalipov et al.,2006) had previously demonstrated that while derived GFP-expressing rhesus pluripotent ESCs injected into 4-to-8-stagefertilized rhesus embryos would incorporate into thetrophectoderm and ICM cells of the expanded blastocystgrown in vitro, efforts to produce a chimeric monkey afterembryo transfer did not succeed.

Interspecies chimeras have been advanced as an alterna-tive process for exploring early human developmentalprocesses and helping address the basic embryology ofhESCs and their potential applications in cell-based therapies(James et al., 2006). The mouse is the best-characterizedmammalian model and perhaps a logical choice for studyinginterspecies chimera: there is an abundance of cheapembryos and recipients; an enormous background literatureon mESCs already exists; and mouse–mouse chimeras arewell established with regards to genetics, strains and proventechniques. Furthermore, mESCs are unencumbered by thematerial transfer agreement (MTA) restrictions currentlyimposed on all NIH Registry hESC lines that explicitly prohibittheir use in animal chimera production. Taken together, theanswers obtained from testing interspecies mouse–NHPchimeras in utero, after chimeric embryo transfer, and invitro might provide new and important information on thedevelopmental potentials of embryonic stem cells. Inaddition, if successful, these intraspecific chimera wouldopen innovative methods for preserving germ lines fromendangered species (Songsasen and Wildt, 2007; Pukazhenthiet al., 2006) as well as specialty biomedical research models(Yang et al., 2008; Chan and Yang, 2009).

Here, we explore mouse–nhpESC chimeras produced withclassic mouse embryo aggregation or blastocyst injectiontechniques (Nagy, 2003). Using GFP-expressing rhesus andbaboon nhpESCs, we demonstrate that nhpESCs associatewith the ICM in expanded mouse blastocysts, but rarely

Page 3: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

30 C. Simerly et al.

proliferate after outgrowth experiments and do not inter-mingle with mouse tissues, as determined by in vitroanalysis. Furthermore, we show that chimeric mouse–nhpESC blastocysts transferred to pseudopregnant mouserecipients produce fetuses but without detectable contribu-tion from the GFP-expressing nhpESCs, as ascertained byimmunohistochemical, PCR and MRI analysis. Collectively,we conclude that interspecies chimera between distantmammals is unfavorable for studying the full pluripotency ofprimate ESCs, lending intellectual support for intraspecificprimate chimeric experimentation.

Results

The rhesus male line nhpESC 2706 was particularly robustfollowing transduction with the EF1α-GFP transgene and couldbe traced in mouse chimera tissues using monkey-specificprimers to the SRY gene. Supplemental Table S1 summarizesthe various stem cell lines employed for preparing injection- oraggregation-produced interspecies mouse chimeras. All of thenhpESCs employed were low passage colonies (range: 7–51) ofgood ESC morphology (Fig. S1A) and maintained their plurip-otent characteristics following transduction with various GFPtransgenes (Fig. S1B), as determined by ‘stemness’ (Fig. S1C;Table S1) and cell surfacemarker expression (Table S1), as wellas their ability to produce teratomas when injected into NOD-

Table 1 Summary of mouse–nhp interspecies chimera fetus pro

[a] [b] [c] [d] [e]

Chimeratype

GFP ESCcell line

Recipienttype

Totalembryo #'stransferred(# of trials)

Totalimplantationsites [IP](% of ET)

Mouse 2Nblastocyst-injection

Rhesusnhp2706

ICR 147 (9) 56 (38)

Rhesusnhp106

ICR 60 (3) 25 (42)

BaboonESC-4

NOD-SCID 32 (4) 8 (25)

Mouse ESC-YFP

ICR 48 (4) 16 (33)

Mouse 2Nembryo-aggregation

Rhesusnhp2706

ICR 39 (4) 37 (95)

Rhesus nhp3006

ICR 18 (1) 18 (100)

Mouse ESC-YFP±bangparticles

ICR 42 (4) 14 (33)

Mouse 4N-injection

Rhesusnhp2706

ICR 9 (1) 9 (100)

Rhesusnhp106

ICR 7 (1) 0

Rhesusnhp3006

ICR 7 (1) 0

a 2/4 recipients died prior to fetal analysis on day E12 post transfer.b 9/25 abnormal fetal tissues demonstrated surviving nhpESC-GFP cec 5/9 reabsorbing or empty sacs demonstrated surviving nhpESC-GFP

SCID strain mice. Additionally, spontaneous differentiation ofGFP-expressing nhpESC colonies in vitro did not silence thetransgene, providing confidence that the primate cells wouldmaintain GFP detection within interspecific chimera construc-tion following embryo transfer (Fig S1D–I). Control intraspe-cific chimeraswere produced by a yellow fluorescent variant ofR1mESCs (7ACS/EYFP; ATCC;Manassas, VA) thatwas germline-competent and stained positive for pluripotency markers(Hadjantonakis et al., 2002).

GFP-expressing Rhesus nhpESC lines (Table 1) used in theaggregation chimera assay produced expanded mouse blasto-cysts with GFP-expressing nhpESC lines 2706 and 3006associated with the blastocyst inner cell mass (ICM) cells. Wetypically combined mouse zona pellucida-free 2-to-8-cellstage embryos with Rhesus or Baboon GFP-expressing nhpESCs(Rhesus: Fig. 1A, arrow) in a depressionwell and cultured themin vitro until the blastocyst stage (see Figs. S6–S8). Afterfixation, confocal optical sectioning (Baboon: Fig. 1B: differ-ential interference contrast (DIC); 1C: Hoechst DNA, blue)demonstrated direct fluorescence detection of baboon GFP-expressing nhpESCs within the ICM (Fig. 1D: GFP, green,arrowheads). Here, no GFP-expressing baboon nhp 2706ESCscells were observed in the outer trophectodermal cells, asdemonstrated with a trophectoderm-specific antibody, CDX2(Fig. 1E: red; arrow, ICM; composite image, Fig. 1F). Weobserved nearly 27% (129/479) of expanded chimera blasto-cysts with GFP-expressing Rhesus or Baboon nhpESCs

duction.

[f] [g] [h] [i]

# Normalfetusesrecovered(% of total IP)

# Abnormalfetusesrecovered(% of total IP)

# ofreabsorbed/empty sacs(% of total IP)

# GFP, YFP orbang particlenormal fetuses(% of total IP)

43 (77) 8 (14) 5 (9) 0

25 (100) 0 0 0

5 (63) 0 3 (38) 0 a

12 (75) 0 4 (25) 5 (31)

7 (19) 19 (51) 11 (30) 0 b

2 (11) 6 (33) 10 (56) 0 b

6 (43) 0 8 (57) 5 (36)

0 0 9 (100) 0 c

0 0 0 0

0 0 0 0

1 reabsorbing embryo expressed nhpESC-GFP cells.lls.cells.

Page 4: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

Figure 1 Generation ofmouse×nhpESC-GFP interspecies aggregation chimera. A: a ‘sandwich’mouse aggregation chimera preparedbymixing a small clump of GFP-expressing nhp2706 ES cells (green; arrow)with two 2-cell mouse embryos in a depressionwell (arrowheads).B–F: confocal image of a fixed interspecies aggregation chimera (B: DIC; and C: DNA) produced with GFP-expressing BabESC-4, showinglocalization of BabESCs (D: green, arrowheads) at themouse ICM [B,C,E: arrows; E: cdx-2, a trophectoderm specificmarker, red] but not inthe outer trophectodermal cells (F: merged imaged). Similar aggregation chimera w Bar=20 μm.

31Interspecies chimera between primate embryonic stem cells and mouse embryos

exclusively in the mouse ICM and another 11% (55/479) withGFP-expressing Rhesus or Baboon nhpESCs in both the ICM andtrophectoderm. The number of GFP-expressing nhpESCsassociated with the mouse ICM was variable, with a majorityshowing 2–5 NHP cells within the ICM of expanded blastocysts.

Time-lapse video microscopy (TLVM) was used to inves-tigate intraspecific and interspecific chimera formation invitro (Figs S6–S8). After 24–48 h of aggregation within adepression well, compacted embryos with adhering mouse or

Rhesus nhp ESCs were collected and prepared for TLVMrecording for development to the expanded blastocyst stage.In intraspecific chimera (mouse×YFP-expressing mouseESCs), video evidence suggested that the adhering mouseYFP-ESC (Fig. S6A–B) would integrate into the mouse duringperiods of blastocoel expansion, often as trophectodermcells were undergoing division. Interestingly, the expandingmouse blastocyst did not collapse during the YFP-ESCincorporation phase, perhaps suggesting that the mouse

Page 5: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

32 C. Simerly et al.

embryo and mouse ESCs share similar cell surface signalingmolecules that could mediate aggregation. This observationwas similar to control blastocysts lacking the zona pellucida(not shown), but distinct from observations reported in otherrodents (Gonzales et al., 1996).

Immunocytochemistry analysis of fixed aggregation chime-ra at the end of the TLVM showed evidence of successful YFP-mouse ESC incorporation near the site of the mouse ICM(Supplemental Fig. S6C–F). We next explored interspecificaggregation chimera formation produced by combining mouseembryos with unlabeled Rhesus nhp 2706 ESC line. Inaggregation chimera that failed to incorporate the nhpESCs,the mouse morula stage rapidly expands into a fully expandedblastocyst. No evidence of breaching the trophectoderm orincorporation of nhp2706 ESCs into the embryo proper wasobserved (Fig. S7). However, incorporation of nhp 2706 ESCsinto the expanding mouse blastocyst appeared to occur justafter a rapid blastocyst collapse (Fig. S8D–E and 8I–J,arrowheads). These cells appeared to move rapidly towardsthe animal pole, or ICM region. Blastocyst collapse is perhapscaused by the breeching and interjection of nhpESCs throughthe tight junction of the adhering outer mouse trophoblastcells.

We transferred mouse 2N embryo aggregation chimeraproduced with GFP-expressing nhp 2706 or 3006 ES cell linesinto ICR strain recipients to investigate fetal contributions invivo (Table 1). Of 57 total aggregation chimera transferred to 5recipients, we observed 55 (96%; Table 1, column e)implantation sites but only 9 (16%; Table 1, column f) normalfetuses. The vastmajority of the tissues recoveredwere eitherabnormal (25 total; 45%; Table 1, column g) or beingreabsorbed (21 total; 38%; Table 1, column h). None of thefetuses recovered demonstrated GFP-expressing nhpESCs(Table 1, column i). Microscopic analysis of the fetal tissuesrecovered from embryos produced with rhesus GFP-expressingnhp2706 cell line showedmany instances of axial abnormalities(head–trunk: Fig. 2A) and delayed fetal development (Fig. 2C)but no detectable GFP-expressing cells (Fig. 2B–D). In severalinstances, surviving GFP-expressing 2706 ES cells wereobserved in reabsorbing implantation tissues (Fig. 2E, bright-field; 2F, GFP, green, arrowheads). Thus, aggregationchimeras with GFP-expressing nhpESCs produced mosaicblastocysts with varying number of GFP cells associated withthemouse ICM and high numbers of abnormal fetuses followingembryo transfer to pseudopregnant recipients.

Next, we explored interspecies chimera after GFP-expressing nhp 2706 or 106 ESCs were injected into theexpandedmouse blastocysts. We first determined the survivalof injected nhp2706 ESCs within the mouse blastocoel niche.Chimeras were produced bymicroinjecting a known number ofGFP-expressing nhp2706 ESCs into expanded mouse blasto-cysts (Fig. S2A), placing the GFP-expressing cells adjacent tothe mouse ICM (Fig. S2B). As shown, within 4–6 h, the re-expanded mouse blastocysts (Fig. S2C) demonstrated GFP-expressing nhp2706 ESCs in the blastocoel, some localized atthe mouse ICM (Fig. S2D: GFP, green). However, only 43% [10/23] of injected blastocysts retained any GFP-positive nhp2706ESCs after 21 h of in vitro culture and fluorescent analysis ofsurviving GFP-nhp2706 ESCs revealed N70% loss of the totalnumber of cells. We then performed embryo transfers ofinjected interspecies chimeric blastocysts to pseudopregnantICR or NOD-SCID mice after using either rhesus or baboon GFP-

expressing ESCs. We observed several implantation sites (ICR:81/207 [39%]; NOD-SCID: 8/32 [25%]; Table 1, columns d–e)and a high percentage of normal E10.5 fetuses at recovery(ICR: 68/81 [84%]; NOD-SCID: 5/8 [63%]; also Fig. S3).However, none of the normal fetuses expressed GFP(Table 1; column i; Fig. S3). Conversely, from 48 embryotransfers using control intraspecific chimeric blastocystsproduced with YFP-expressing mESCs, 16 implantation sites(33%; Table 1, column e) and 12 normal fetuses (75%; Table 1,column f) were recovered, with 5 fetuses expressing YFP (31%;Table 1, column i; see also, Fig. S3). Microscopic analysis ofinjection chimeric embryos produced with GFP-expressingRhesus nhp 2706 ESCs (Fig. 3) or GFP-BabESC4 cell lines(Fig. S4) in either ICR or NOD-SCID recipients demonstrated noGFP expression in the tissues of recovered normal fetuses(Rhesus 2706: Fig. 3A–B; BabESC-4: Fig. S4A–B). Variousabnormal embryos were largely negative for GFP detectionalso (Rhesus 2706: Fig. 3A–B), although occasional GFP ‘dots’were observed in some recovered abnormal tissues (Rhesus2706: Fig. 3E, brightfield; 3F, GFP, green, arrowheads; Fig.S4E, brightfield; Fig. S4F, GFP, green, arrowhead; BabESC-4:Fig. S4C, brightfield; S4D: GFP, green, arrowheads). Mouseintraspecific embryos produced with YFP-expressing mESCsand transferred to pseudopregnant recipients showed exten-sive fluorescence throughout the E10.5 day fetus (Fig. 3G–H;YFP, yellow).

Mouse–nhpESC chimeras were also prepared using themouse tetraploid complementation assay (Nagy, 2003). Weelectrofusedmouse 2-cell embryos to produce 4N embryos andpermitted these to develop to the expanded blastocyst stagebefore injecting them with GFP-expressing nhpESCs andperforming embryo transfers to ICR pseudopregnant recipi-ents. Harvest of fetal material around E12.5 showed highimplantation sites with chimera prepared with nhpESC 2706ESCs (Table 1, columne), but no normal fetal development andmostly necrotic or reabsorbing implantation sites uponsacrifice (Table 1, column h).

To analyze mouse–nhpESC chimeras at the cellular level, afew normal E12.5 fetuses were selected for immunohisto-chemical analysis (Fig. S5). We counterstained 10 μm sectionswith anti-GFP antibody to compare with any detected GFPexpression. Preliminary analysis of ectoderm (spinal cordtissue: Fig. S5A1),mesoderm (pericardial tissue: Fig. S5B1) andendoderm (urogenital tissue; Fig. S5C1) layers in an interspe-cies fetus suggested extensive survival of GFP-expressing cellsthat co-localized precisely with anti-GFP staining (Fig. S5A2,S5B2, and S5C2). However, control tissue sections from afertilized E12.5 day mouse embryo suggested extensive auto-fluorescence following fluorescein and rhodamine excitationin a variety of tissues, rendering fluorescent analysis unreliable(fluorescein excitation: Fig. S5A3, S5B3, and S5C3; rhodamineexcitation: Figs. S5A4, S5B4, and S5C4). Efforts to control forendogenous fluorescence by using various blocking agentsprior to application of primary and secondary antibodies werenot successful (our unpublished data).

On selected interspecies embryos produced with GFP-expressing rhesus nhp2706 ESCs (a male line), we explored ifSRY and GFP DNA could be detected by PCR analysis (Fig. 4).For a positive control, we used DNA isolated from atransgenic male monkey carrying the GFP transgene (ANDi)(Chan et al., 2000), demonstrating the detection of SRY DNA(Fig. 4, lane 3) and GFP DNA (Fig. 4, lane 14). However, no

Page 6: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

Figure 2 Developmental abnormalities in interspecies aggregation chimera at E10.5 prepared with GFP-expressing nhp 2706 ESCs.A–B: head–trunk axial deformity (A: BF; arrowheads, A: anterior head region; P: posterior trunk region) in an aggregation chimera. NoGFP expression was observed (B: green). C–D: severely delayed embryonic development (C: BF), but no discernible GFP expression(D: green). The tail region was slightly damaged during dissection from the decidual sac. E–F: an implantation site without a definableembryo (E: BF) but with a few GFP expressing cells (F: green, arrowheads). BF: brightfield optics; A: anterior head region; P: posteriortrunk region. Bars=500 μm.

33Interspecies chimera between primate embryonic stem cells and mouse embryos

DNA from the embryonic tissues of these interspecieschimeras produced positive bands with primers from eitherSRY (Fig. 4, lanes 4–6) or GFP (Fig. 4, lanes 9–13), suggestingthat no nhpESCs had survived in the developing mousefetuses. Analysis of an interspecies chimeric blastocystoutgrowth produced with GFP-expressing nhp2706 ESCs alsodid not detect SRY DNA after 1 month in culture (Fig. 4, lane2). This particular colony did not demonstrate GFP expres-sion in surviving cells after a few days of culturing in vitro.

Finally, we investigated GFP-expressing nhpESC survival,proliferation and integration with mouse cells in vitrofollowing blastocyst outgrowth on sterile coverslips

(Fig. 5). Chimeric injection blastocysts outgrown for 3 daysshowed that GFP-expressing cells remained largely clusteredtogether without significant intermixing with mouse cells(Fig. 5A–C). Likewise, aggregation chimeric blastocystoutgrown for 17 days in vitro (Fig. 5D–F) demonstratedthat while the GFP-expressing nhpESCs proliferated over the2 weeks in culture, the nhpESCs did not integrate into themouse ICM cellular area (Fig. 5D–F, * indicates mousedifferentiated cells derived from the mouse ICM). Regard-less, the survival of pluripotent, GFP expressing nhpESCs inmouse chimeric blastocyst outgrowths were low (~2.5%)overall.

Page 7: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

Figure 3 Chimeric blastocyst injection embryos at E10.5 day post embryo transfer. A–B: Normal embryo produced from amouse×GFP nhp2706-ESC injection chimera (A: BF), but without GFP expression (B: green). C–D: An abnormal mouse embryo (C: BF)derived from the transfer of a mouse×GFP-expressing nhp2706-ESCs injection chimeric. No GFP expressing cells are seen in thedisorganized tissue (D: green, GFP). E–F: a reabsorbing mouse embryo (E: BF) derived from a mouse×GFP-expressing nhp2706-ESCsinjection chimeric. A few GFP-expressing cells are observed in the fetus (F: green, arrowheads). G–H: Control chimeric embryoderived from a mouse×YFP-mouse ESCs blastocyst transfer into an ICR recipient. The normal embryo (I: BF) expresses YFP in manytissues (J: YFP). BF: bright field; GFP: green fluorescent protein. All embryo transfer were performed in ICR recipients. All chimerawere produced with GFP-expressing rhesus 2706 male ESC line. Bar=500 μm.

34 C. Simerly et al.

Discussion

The contribution of ESCs and other PSCs to chimeric offspringresulting in germ-line transmission is the most stringentassay for demonstrating biological pluripotency (reviewed by

Behringer, 2007). This chimera assay has resulted in significantinsights into the various categories of PSCs, even with mice,since embryonal carcinoma, embryonic germ, ESCs and PSCsgenerated by induced pluripotency all pass this test, whereasstem cells from epiblasts do not (Tesar et al., 2007; Brons

Page 8: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

Figure 4 PCR analysis of SRY (Left) and GFP (Right) DNA in embryos derived from mouse blastocyst injected with GFP nhp ESC 2706male line. Lane 1: DNA marker; lane 2: chimeric outgrowth without GFP positive cell expression; lane 3: positive DNA control(transgenic monkey cells from ANDi); lanes 4–6: embryos derived from an injection chimera attempt (mouse×GFP nhpESC 2706 cellline); lane 8: DNA marker; lanes 9–13: embryos derived from an injection chimera (mouse×GFP nhpESC 2706 cell line); lane 14:positive DNA control (ANDi cells); lane 15: blank.

35Interspecies chimera between primate embryonic stem cells and mouse embryos

et al., 2007). Epiblast SCs, inwhich Lif signalingwas introducedtransgenically (Bao et al., 2009), were shown to have regainedthe ability to participate in chimeric development and

Figure 5 Interspecies chimeric outgrowth. A–C: Confocal imaging oESCs at day 3 post-outgrowth. The GFP-nhpESCs remain clusteredB: Hoechst DNA; C: merged image. D–F: Sequential fluorescentaggregation chimeric blastocyst produced with male GFP nhp2706 Eonto the MEF feeders with extensive GFP nhpESCs at the mouse ICM (D(E: *) are distinctly separate from the expanding GFP-nhpESCs (Eoutgrowth, rapid proliferation of the GFP-expressing nhpESCs along ais observed, with little intermixing of the mouse: monkey ESCs. Mag

transmit to the germ-line, demonstrating that the loss of thissignaling cascade during post-implantation development re-sults, in part, with this diminishment of pluripotency.

f a mouse blastocyst injected with male GFP-expressing nhp2706together (A: green) without intermixing with mouse ICM cells.and Hoffman Modulation Contrast (HMC) images of a mouseSCs. At day 2 post outgrowth, the chimeric blastocyst attaches: green). On day 8 post-outgrowth (E), the expanding mouse ESCs: green; inset, details of GFP-nhpESCs). On day 17 days post-distinct border of the largely differentiated mouse ESC colony (*)=100×; Bar=20 μm.

Page 9: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

36 C. Simerly et al.

Chimera have been generated in the lab exclusively asinterspecific chimera between mouse species (Rossant andFrels, 1980) and recently rats (review by Iannaccone andJacob, 2009). Even within the rodent family, intergenericchimerabetweenmice and voles did not succeed (Mystkowska,1975a, 1975b). In domestic species, intergeneric chimerawerefirst generated between sheep and goat embryos more thansixty years ago (Warwick and Berry, 1949) while interspecificchimera between European and indigenous Asian cattle havealso been generated (Williams et al., 1990). Pregnanciesgenerated by the ovine–caprine intergeneric chimera succeedto term but only at low frequencies (Gustafson et al., 1993;Jaszczak et al., 1999) demonstrating the loss of chimeraproportions as these animals age post-natally.

Among the several rationales for this study, four are mostprominent. First, it is important to understand the develop-mental biology of embryonic stem cells, as well as other linesnow classified within the constellation of pluripotent stemcells. While the fundamental science of this field is on firmfoundations with the decades of confirmed reports usingmouse ESCs, results in other species, including humans andother primates, rest on less sure footings. Related to this point,the enormous expansion of the PSC field as well asunderstandable regulatory constraints on using hESC chimeraassays has resulted in the proliferation of numerous alternativepluripotency assays with various degrees of leniency. Indeed,if pluripotency is viewed as a scale in which assays are ratedfrom greatest stringency to most permissive, then germ-linetransmission in tetraploid embryo complementation would beconsidered at the most reliable. Perhaps less stringent wouldbe fertilized embryo chimera. Owing to the interest in humanESCs, in which only one group has reported chimeric assays(James et al., 2006), teratoma assays serve as the moststringent test for pluripotency in which tissues from the threegerm layers are examined. Notwithstanding the practicality ofthese teratoma assays, organogenesis and patterning arechaotic and the extent of germ layer contributions is rarelyquantified. Embryoid bodies and in vitro differentiation, eitherspontaneous or directed, are perhaps mid-scale on thispluripotency assay ruler. The detection of pluripotencymarkers by fluorescence (i.e., Oct-4, NANOG, SSEAs, andTra-1-antigens) is problematic due to problems of autofluor-escence, cross-reactivity as well as non-specific expression.RT-PCR is extraordinarily sensitive which forces questionsabout whether minute numbers of contaminating cells mightgenerate misleading results. Notwithstanding the power oftranscriptional analysis and its potential contributions forsystem biology, the reliability of these in silica approaches forunequivocal demonstrations of biological pluripotency re-mains to be confirmed. Consequently, the prime rationale forthis investigation was to determine in a relevant biologicalassay the post-implantation potential of nhpESCs in murinechimera.

Secondly, the field of pluripotency is rapidly influencing thedesign of futuremedical approaches. Withmice, few concernsare raised as to whether a transgenic insertion of GFP mightinfluence the outcome of experiments, thus the importance ofmore reliably understanding various increases in perturbationsas fundamental studies move towards clinical applications.Against this background, James et al. (2006) conducted acomplicated set of experiments in which they first establisheda unique hESC line which was free from the MTA (material

transfer agreements) of the traditional stem cell supplies,since those MTAs prohibit the introduction of hESCs into thereproductive systems of mammals or the combination of hESCswith embryos for reproductive purposes. Also, they were ableto conduct their investigations without federal funding re-strictions that preclude these types of experiments. This studysuggested that human ESCs introduced into mouse blastocystsby either aggregation or blastocyst injection survived withinthe mouse ICM niche and proliferated into differentiatedhuman derivatives. Furthermore, the human ESCs weredescribed as integrated into early embryonic mouse tissuesfollowing embryo transfer to pseudopregnant females. Not-withstanding heroic efforts in performing these investigations,questions remain regarding whether the introduced hESCsproliferated and participated in post-implantation develop-ment. Perhaps they were ‘bystanders’ surviving on thesidelines and swept up in the morphogenetic migrations.Questions have been raised as to whether the foci detected byfluorescence might even have been adventitious. Perhaps thehESC line generated from anonymously-donated clinically-discarded specimens might have been subprime owing to itsorigins. Consequently, we undertook these studies usingembryos generated by fertile pedigreed primates for theexpress purpose of generating top-quality ESC lines with thebest chances for full biological pluripotency.

Third, beyond fundamental and preclinical significance,the importance of chimeric assays using nonhuman primatesextends into the realm of invaluable research resources.Investigations using nonhuman primates are expensive andcumbersome, yet important to bridge the gap from funda-mental discoveries in mouse models to clinical investigations.Were PSCs from NHPs to turn out useful in generating chimericoffspring, a significant number of investigations could beperformed in vitro with only the last confirmatory studiesconducted on specialty primates, as is the case withmice. Theopportunity to modify primate research resource require-ments using chimera would be rather significant, especially asinnovative research models (reviewed by Schatten andMitalipov, 2009) are emerging, including transgenic primates(Yang et al., 2008; Chan et al., 2000), NHPs with discordantmitochondrial and nuclear genetics (Tachibana et al., 2009),and perhaps reproductive clones soon.

Fourth, bioethical considerations regarding chimera be-tween human cells and animal embryos, related concernsinvolving transfer of human nuclei into enucleated animaloocytes [cybrids], as well as actual hybrids are topics of activedebate (Chapman and Hiskes, 2008; St John and Lovell-Badge,2007). To help ground these bioethical conversations on afirmer foundation, this investigation, using nonhuman primatestem cells chimerized into mouse embryos, was designed toaddress the biological feasibilities of this assay. It is importantto note that in contrast to the human ESCs available, theseprimate lines were all generated from fertile pedigreedprimates where the best quality embryos were selected fromESC derivations. Should chimeric fetuses or animals begenerated using pluripotent stem cells and either interspecificor intraspecific animal embryos, then the biological founda-tion for this experimental manner of reproduction wouldunderscore the recent calls to prohibit hiPSCs for reproductivecloning (Lo et al., 2010).

Here, we demonstrate that chimeric embryos generatedby combining mouse embryos with nhpESCs from either

Page 10: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

37Interspecies chimera between primate embryonic stem cells and mouse embryos

rhesus or baboons are not detected after implantation, evenwhen the primate cells localize to the ICM in both aggregatedand injected embryos. Aggregation chimera display numer-ous nhpESCs at the mouse ICM but fetal development afterembryo transfers is significantly impaired (Fig. 2; Table 1),while chimera generated by blastocyst injection have fewernhpESC within ICM but the surviving fetuses are moredevelopmentally normal (Fig. 3; Table 1). Consequently,we suggest that these interspecific chimeric embryos mayhave limited pre-clinical utility for analyzing the pluripotentstatus and developmental capabilities of primate ESC.

Autofluorescence in mouse tissues raises significantconcerns of ‘false positive’ interpretations. In Fig. 6, wepresent an immunohistochemistry analysis of fetal tissuesboth before and after introduction of the anti-GFP antibody.The danger of premature enthusiasm from extensive mousefetal autofluorescence is quite high (Fig. 4). However, thesurvival and/or participation of monkey ESCs in fetal micewere either below detection sensitivity or perhaps precludedby biological incompatibilities. PCR analysis using specificprimers to the monkey SRY gene or GFP did not detect thepresence of male GFP-expressing nhpESCs in the tissues ofmid-stage embryos (Fig. 5). Additionally, we exploredtracing ESC participation in developing mouse chimerausing high magnetic field MRI microscopy. Mouse YFP-expressing ESCs pre-labeled with Bangs™ beads, superpar-amagnetic microparticles detectable by MRI (Shapiro et al.,2004), were used to prepare mouse chimeric blastocystsfollowing aggregation or injection into mouse blastocysts.However, despite 38% mouse chimeric production followingtransfer to pseudopregnant females (Table 1), as assayed byGFP expression, we could not detect the Bangs beads by MRImicroscopy as have been previously reported (Shapiro et al.,2004) imaging, perhaps owing to particle levels below MRIdetectable thresholds within the tissues. Newer methodscoupling transgene reporters with ferritin may be moresuitable for investigating individual cell contribution tochimeric tissue and organs (Ahrens et al., 2006; Mills andAhrens, 2009; Genove et al., 2005).

It is tempting to speculate that differences in the cellularadhesiveness between rodents and primates preclude theirmigration during gastrulation and beyond. It appears thatESCs prefer to adhere with cells of their own species andperhaps this specific–specific differential adhesion accountsfor the results here. When outgrowths of these interspecificnhpESC–mouse chimeras are established, the murine cellsappear to grow separately from the growing nhpESC ones,i.e. the surviving colonies do not intermix, but self-select to‘like’ cells (Fig. 6). Perhaps the adhesive requirements forcells to remain attached during the morphogenetic move-ments at gastrulation block nhpESC contribution to thedeveloping fetus because their association with the murinecells is too weak.

Perhaps cell cycle differences between primate androdent pluripotent stem cells preclude primate ESCs fromparticipation in development after implantation. The timecourse of development also differs significantly betweenrodents and primates. Blastocysts develop in mice within3.5–4 days, whereas human blastocysts require 5–6 days andnhp primates a week or more. Perhaps differences in cellcycle influence the relative proliferation of nhpESCs withinthe differently timed mouse embryo. It is also worth noting

that whereas mouse gestation is around three weeks, rhesusand baboons require over a half-year.

Recent evidence has shown that human ESCs have similarcharacteristics to mouse epiblast stem cells (EpiSCs) ratherthan mouse ESCs. Like hESCs, mouse EpiSCs demonstratesimilar dependence on bFGF/Activin signaling, grow inflatter colony morphologies with slower growth patternscompared to mESCs, and show similarities in X-chromosomeinactivation (Tesar et al., 2007; Brons et al., 2007). Also,since mouse EpiSCs do not typically produce chimera inintraspecific chimera assays, it may be that nonhumanprimate ESCs will also be poor candidates for chimeraproduction in monkey: monkey chimera attempts. However,methods are now being discovered that permit intraconvert-ibility of human ESCs into more murine ESC-like states(Nichols and Smith, 2009; Buecker et al., 2010; Kerr andCheng, 2010; Xu et al., 2010). Hanna et al. (Hanna et al.,2010) recently demonstrated that hESCs can be repro-grammed into a more mouse ESC like states with regards togene expression profiles, X chromosome inactivation infemale lines, and Lif/Stat3 signaling. While human PSCscannot be used for intraspecific chimera attempts, repro-gramming nonhuman primate ESCs into this naïve state canbe tested to determine if it improves interspecific chimeraresults for the demonstration of full pluripotentiality as wellas for enhanced biomedical utility of these preclinicalresearch resources.

Conclusions

ESCs from both baboons and rhesus integrate into the ICM ofmouse blastocysts in both aggregation and injection chimera,but they are lost after implantation. This suggests thatinterspecies chimera may have limited pre-clinical diagnosticutility for determining the developmental potentials of cellsfrom primates. Regardless, the likelihood that primate ESCsmay participate in chimeric development in intraspecificembryos remains both with currently available nhpESCs andperhaps more successfully with naïve nhpESCs.

Material and methods

Mice

Female F1 B6D2F1 mice (Harlan Sprague Dawley, Indianap-olis, IN) were hormonally superstimulated, bred to fertilemales, and collected as described previously (Simerly andSchatten, 1993). ICR or NOD-SCID strain females mated tovasectomized ICR males (Harlan) produced pseudopregnantfemale recipients for embryo transfers (ETs) (Nagy, 2003).

ESC lines

Mouse YFP-expressing embryonic stem cells (mESCs) wereobtained commercially (ATCC; Manassas, VA) and cultured asdescribed (Hadjantonakis et al., 2002). Nonhuman primateembryonic stem cell lines were derived and maintained onmitomycin-C inactivated primary mouse embryonic fibro-blasts (MEFs) on 0.1% gelatin-coated dishes as previouslydescribed (Navara et al., 2007a; Simerly et al., 2009).

Page 11: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

38 C. Simerly et al.

Supplemental Table S1 list the pluripotency marker charac-teristics and teratoma data outcomes for 3 transducedRhesus lines (nhp 2706; nhp 3006; and nhp 106) and 1 baboonline (BabESC-4) employed for this study. All NhpESCs colonieswere mechanically passaged weekly with culture mediumchanged every 48 hours. GFP transduction of nhpESC lines isdescribed in the Supplemental data.

Interspecies chimera production

Colonies of YFP-mESCs or GFP-nhpESCs were briefly treatedwith 0.05% trypsin-EDTA (45 s) at 37 °C,mechanically scraped,and washed once. Aggregation chimeras were prepared bycombining zona pellucida-free mouse embryos (4-to-8 cellstage) with small clumps (~10–50 cells) of transduced mouseor monkey ESCs in depression wells (Nagy, 2003) using theappropriate stem cell media. Aggregations were grown untilthe expanded blastocyst stage and analyzed for incorporationat the mouse blastocyst ICM using attenuated fluorescentexposure (b5 s) on a Nikon TE-300 inverted microscope.

For injection chimeras, small clumps of transducedmouse ormonkey ESCs were microinjected into expanded mouse blasto-cysts using beveled pipettes (17 μm; Humagen, Charlottesville,VA). Injected mouse blastocysts were recovered 6 h before ETsinto pseudopregnant recipients. Methods for producing tetra-ploidmouse chimeric embryos are provided in the SupplementalData.

Outgrowths

The zona pellucida of aggregation- or injection-producedexpanded chimeric mouse blastocysts were removedwith acidTyrode's (Specialty Media, Millipore Corporation, Bedford,Mass), recovered for 30 min, and then plated onto MEFs inrhesus stem cell media (Navara et al., 2007b). Media changeswere performed every 48 h and images taken by inverted HMCand fluorescence optics.

Embryo transfers and fetal recoveries

Chimeric blastocysts produced by aggregation or injectionmethods were transferred into the uterine horns of day 2.5pseudopregnant ICR or NOD-SCID females using aseptictechniques (Nagy, 2003). Analysis of fetal development inrecipients was performed between E12–17.5 days. Bright-field and fluorescent photographs were taken of excisedfetuses using a Nikon Digital Sight DS-5MC CCD on a NikonSMZU dissecting scope (Nikon USA, Melville, NY). Digitalimages were archived using MediaView software (MolecularDevices, Sunnyvale, CA). Abnormal embryos were given GD(growth disorganization) scores as described Poland et al.(Poland et al., 1981).

Immunocytochemistry, Immunohistochemistry, PCR, andTime-lapse Video Microscopy (TLVM) details are described inthe Supplemental data.

Acknowledgments

We are grateful to Tony Battelli (MWRI&F) for animal caresupport and thank Angela Palermo Lauff for the editorial

assistance. We also thank Drs. R. Pedersen (CambridgeUniversity) and P. Donovan (University of California—Irvine)for the advice. The support of this research to GS by grantsfrom the National Institutes of Health (HD047675; andRR013632) is gratefully acknowledged.

Appendix A. Supplementary data

Supplementary data to this article can be found online atdoi:10.1016/j.scr.2011.03.002.

References

Ahrens, E.T., Srinivas, M., Capuano, S., Simhan, H.N., Schatten, G.P.,2006. Magnetic resonance imaging of embryonic and fetaldevelopment in model systems. Methods Mol. Med. 124, 87–101.

Bao, S., Tang, F., Li, X., Hayashi, K., Gillich, A., Lao, K., Surani, M.A.,2009. Epigenetic reversion of post-implantation epiblast topluripotent embryonic stem cells. Nature 461, 1292–1295.

Behringer, R.R., 2007. Human–animal chimeras in biomedicalresearch. Cell Stem Cell 1, 259–262.

Boyd, A.S., Wu, D.C., Higashi, Y., Wood, K.J., 2008. A comparison ofprotocols used to generate insulin-producing cell clusters frommouse embryonic stem cells. Stem Cells 26, 1128–1137.

Brons, I.G., Smithers, L.E., Trotter, M.W., Rugg-Gunn, P., Sun, B.,de Sousa Lopes, S.M. Chuva, Howlett, S.K., Clarkson, A.,Ahrlund-Richter, L., Pedersen, R.A., Vallier, L., 2007. Derivationof pluripotent epiblast stem cells from mammalian embryos.Nature 448, 191–195.

Buecker, C., Chen, H.H., Polo, J.M., Daheron, L., Bu, L., Barakat, T.S.,Okwieka, P., Porter, A., Gribnau, J., Hochedlinger, K., Geijsen, N.,2010. A murine ESC-like state facilitates transgenesis andhomologous recombination in human pluripotent stem cells. CellStem Cell 6, 535–546.

Chan, A.W., Yang, S.H., 2009. Generation of transgenic monkeyswith human inherited genetic disease. Methods 49, 78–84.

Chan, A.W., Luetjens, C.M., Schatten, G.P., 2000. Sperm-mediatedgene transfer. Curr. Top. Dev. Biol. 50, 89–102.

Chapman, A., Hiskes, A.L., 2008. Unscrambling the eggs: cybridresearch through an Embryonic Stem Cell Research OversightCommittee (ESCRO) lens. Am. J. Bioeth. 8, 44–46.

Conley, B.J., Denham, M., Gulluyan, L., Olsson, F., Cole, T.J.,Mollard, R., 2005. Mouse embryonic stem cell derivation, andmouse and human embryonic stem cell culture and differentia-tion as embryoid bodies. Curr. Protoc. Cell Biol. 23 Chapter, Unit23 2.

Daley, G.Q., Ahrlund Richter, L., Auerbach, J.M., Benvenisty, N.,Charo, R.A., Chen, G., Deng, H.K., Goldstein, L.S., Hudson, K.L.,Hyun, I., Junn, S.C., Love, J., Lee, E.H., McLaren, A., Mummery,C.L., Nakatsuji, N., Racowsky, C., Rooke, H., Rossant, J.,Scholer, H.R., Solbakk, J.H., Taylor, P., Trounson, A.O., Weiss-man, I.L., Wilmut, I., Yu, J., Zoloth, L., 2007. Ethics. The ISSCRguidelines for human embryonic stem cell research. Science 315,603–604.

Dighe, V., Clepper, L., Pedersen, D., Byrne, J., Ferguson, B.,Gokhale, S., Penedo, M.C., Wolf, D., Mitalipov, S., 2008.Heterozygous embryonic stem cell lines derived from nonhumanprimate parthenotes. Stem Cells 26, 756–766.

Eakin, G.S., Behringer, R.R., 2003. Tetraploid development in themouse. Dev. Dyn. 228, 751–766.

Elkabetz, Y., Panagiotakos, G., Al Shamy, G., Socci, N.D., Tabar, V.,Studer, L., 2008. Human ES cell-derived neural rosettes reveal afunctionally distinct early neural stem cell stage. Genes Dev. 22,152–165.

Page 12: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

39Interspecies chimera between primate embryonic stem cells and mouse embryos

Evans, M., 2005. Embryonic stem cells: a perspective. NovartisFound. Symp. 265, 98–103 discussion 103-6, 122-8.

Evans, M.J., Kaufman, M.H., 1981. Establishment in culture ofpluripotential cells from mouse embryos. Nature 292, 154–156.

Fujimoto, A., Mitalipov, S.M., Clepper, L.L., Wolf, D.P., 2005.Development of a monkey model for the study of primategenomic imprinting. Mol. Hum. Reprod. 11, 413–422.

Fujimoto, A., Mitalipov, S.M., Kuo, H.C., Wolf, D.P., 2006. Aberrantgenomic imprinting in rhesus monkey embryonic stem cells. StemCells 24, 595–603.

Genove, G., DeMarco, U., Xu, H., Goins, W.F., Ahrens, E.T., 2005. Anew transgene reporter for in vivo magnetic resonance imaging.Nat. Med. 11, 450–454.

Gonzales, D.S., Boatman, D.E., Bavister, B.D., 1996. Kinematics oftrophectodermprojections and locomotion in theperi-implantationhamster blastocyst. Dev. Dyn. 205, 435–444.

Gustafson, R.A., Anderson, G.B., BonDurant, R.H., Sasser, G.R.,1993. Failure of sheep–goat hybrid conceptuses to develop toterm in sheep–goat chimaeras. J. Reprod. Fertil. 99, 267–273.

Hadjantonakis, A.K., Macmaster, S., Nagy, A., 2002. Embryonic stemcells and mice expressing different GFP variants for multiple non-invasive reporter usage within a single animal. BMC Biotechnol.2, 11.

Hanna, J., Cheng, A.W., Saha, K., Kim, J., Lengner, C.J., Soldner,F., Cassady, J.P., Muffat, J., Carey, B.W., Jaenisch, R., 2010.Human embryonic stem cells with biological and epigeneticcharacteristics similar to those of mouse ESCs. Proc. Natl. Acad.Sci. U.S.A 107, 9222–9227.

Hyun, I., Taylor, P., Testa, G., Dickens, B., Jung, K.W., McNab, A.,Robertson, J., Skene, L., Zoloth, L., 2007. Ethical standards forhuman-to-animal chimera experiments in stem cell research.Cell Stem Cell 1, 159–163.

Iannaccone, P.M., Jacob, H.J., 2009. Rats! Dis. Model. Mech. 2,206–210.

James, D., Noggle, S.A., Swigut, T., Brivanlou, A.H., 2006.Contribution of human embryonic stem cells to mouse blasto-cysts. Dev. Biol. 295, 90–102.

Jaszczak, K., Parada, R., Guszkiewicz, A., 1999. Cytogenetic studyof some tissues and age-related changes in cell proportions in agoat–sheep chimera. Cytogenet. Cell Genet. 84, 55–57.

Kawasaki, H., Suemori, H., Mizuseki, K., Watanabe, K., Urano, F.,Ichinose, H., Haruta, M., Takahashi, M., Yoshikawa, K.,Nishikawa, S., Nakatsuji, N., Sasai, Y., 2002. Generation ofdopaminergic neurons and pigmented epithelia from primate EScells by stromal cell-derived inducing activity. Proc. Natl. Acad.Sci. U S A 99, 1580–1585.

Kerr, C.L., Cheng, L., 2010. Multiple, interconvertible states ofhuman pluripotent stem cells. Cell Stem Cell 6, 497–499.

Lallemand, Y., Brulet, P., 1990. An in situ assessment of the routesand extents of colonisation of the mouse embryo by embryonicstem cells and their descendants. Development 110, 1241–1248.

Lensch, M.W., Schlaeger, T.M., Zon, L.I., Daley, G.Q., 2007.Teratoma formation assays with human embryonic stem cells: arationale for one type of human–animal chimera. Cell Stem Cell1, 253–258.

Liu, H., Zhu, F., Yong, J., Zhang, P., Hou, P., Li, H., Jiang, W., Cai,J., Liu, M., Cui, K., Qu, X., Xiang, T., Lu, D., Chi, X., Gao, G., Ji,W., Ding, M., Deng, H., 2008. Generation of induced pluripotentstem cells from adult rhesus monkey fibroblasts. Cell Stem Cell 3,587–590.

Lo, B., Parham, L., Alvarez-Buylla, A., Cedars, M., Conklin, B.,Fisher, S., Gates, E., Giudice, L., Halme, D.G., Hershon, W.,Kriegstein, A., Kwok, P.Y., Wagner, R., 2010. Cloning mice andmen: prohibiting the use of iPS cells for human reproductivecloning. Cell Stem Cell 6, 16–20.

Martin, G.R., 1981. Isolation of a pluripotent cell line from earlymouse embryos cultured in medium conditioned by teratocarci-noma stem cells. Proc. Natl. Acad. Sci. U S A 78 7634-1638.

Meissner, A., Wernig, M., Jaenisch, R., 2007. Direct reprogrammingof genetically unmodified fibroblasts into pluripotent stem cells.Nat. Biotechnol. 25, 1177–1181.

Mills, P.H., Ahrens, E.T., 2009. Enhanced positive-contrast visual-ization of paramagnetic contrast agents using phase images.Magn. Reson. Med. 62, 1349–1355.

Mitalipov, S.M., 2006. Genomic imprinting in primate embryos andembryonic stem cells. Reprod. Fertil. Dev. 18, 817–821.

Mitalipov, S., Kuo, H.C., Byrne, J., Clepper, L., Meisner, L.,Johnson, J., Zeier, R., Wolf, D., 2006. Isolation and character-ization of novel rhesus monkey embryonic stem cell lines. StemCells 24, 2177–2186.

Mitalipov, S., Clepper, L., Sritanaudomchai, H., Fujimoto, A., Wolf,D., 2007. Methylation status of imprinting centers for H19/IGF2and SNURF/SNRPN in primate embryonic stem cells. Stem Cells25, 581–588.

Mizuseki, K., Sakamoto, T., Watanabe, K., Muguruma, K., Ikeya, M.,Nishiyama, A., Arakawa, A., Suemori, H., Nakatsuji, N.,Kawasaki, H., Murakami, F., Sasai, Y., 2003. Generation ofneural crest-derived peripheral neurons and floor plate cellsfrom mouse and primate embryonic stem cells. Proc. Natl. Acad.Sci. U S A 100 5828-2833.

Mystkowska, E.T., 1975a. Preimplantation development in vivo andin vitro in bank voles, Clethrionomys glareolus, treated withPMSG and HCG. J. Reprod. Fertil. 42, 287–292.

Mystkowska, E.T., 1975b. Development of mouse-bank vole inter-specific chimaeric embryos. J. Embryol. Exp. Morphol. 33,731–744.

Nagy, Z.P., 2003. Micromanipulation of the human oocyte. Reprod.Biomed. Online 7, 634–640.

Nagy,A.,Gocza, E., Diaz, E.M., Prideaux, V.R., Ivanyi, E.,Markkula,M.,Rossant, J., 1990. Embryonic stem cells alone are able to supportfetal development in the mouse. Development 110, 815–821.

Nakatsuji, N., Nakajima, F., Tokunaga, K., 2008. HLA-haplotypebanking and iPS cells. Nat. Biotechnol. 26, 739–740.

Navara, C.S., Redinger, C., Mich-Basso, J., Oliver, S., Ben-Yehudah,A., Castro, C., Simerly, C., 2007a. Derivation and characteriza-tion of nonhuman primate embryonic stem cells. Curr. Protoc.Stem Cell Biol. 1 Chapter, Unit 1A 1.

Navara, C.S., Mich-Basso, J.D., Redinger, C.J., Ben-Yehudah, A.,Jacoby, E., Kovkarova-Naumovski, E., Sukhwani, M., Orwig, K.,Kaminski, N., Castro, C.A., Simerly, C.R., Schatten, G., 2007b.Pedigreed primate embryonic stem cells express homogeneousfamilial gene profiles. Stem Cells 25, 2695–2704.

Nichols, J., Smith, A., 2009. Naive and primed pluripotent states.Cell Stem Cell 4, 487–492.

Okita, K., Ichisaka, T., Yamanaka, S., 2007. Generation of germline-competent induced pluripotent stem cells. Nature 448, 313–317.

Okita, K., Nakagawa, M., Hyenjong, H., Ichisaka, T., Yamanaka, S.,2008. Generation of mouse induced pluripotent stem cellswithout viral vectors. Science 949–953.

Poland, B.J., Miller, J.R., Harris, M., Livingston, J., 1981.Spontaneous abortion. A study of 1,961 women and theirconceptuses. Acta. Obstet. Gynecol. 5–32.

Pukazhenthi, B., Comizzoli, P., Travis, A.J., Wildt, D.E., 2006.Applications of emerging technologies to the study and conser-vation of threatened and endangered species. Reprod. Fertil.Dev. 18, 77–90.

Rajesh, D., Chinnasamy, N., Mitalipov, S.M., Wolf, D.P., Slukvin, I.,Thomson, J.A., Shaaban, A.F., 2007. Differential requirementsfor hematopoietic commitment between human and rhesusembryonic stem cells. Stem Cells 25, 490–499.

Riazi, A.M., Kwon, S.Y., Stanford, W.L., 2009. Stem cell sources forregenerative medicine. Methods Mol. Biol. 482, 55–90.

Roberts, R.M., 2005. Embryo culture conditions: what embryos likebest. Endocrinology 146, 2140–2141.

Rossant, J., 2001. Stem cells in the mammalian blastocyst. HarveyLect. 97, 17–40.

Page 13: Interspecies chimera between primate embryonic stem cells ... · ate into all three germ layers in the adult body (Evans and Kaufman, 1981; Martin, 1981; Thomson et al., 1998). These

40 C. Simerly et al.

Rossant, J., Frels, W.I., 1980. Interspecific chimeras in mammals:successful production of live chimeras between Mus musculusand Mus caroli. Science 208, 419–421.

Rugg-Gunn, P.J., Ferguson-Smith, A.C., Pedersen, R.A., 2005a.Human embryonic stem cells as a model for studying epigeneticregulation during early development. Cell Cycle 4, 1323–1326.

Rugg-Gunn, P.J., Ferguson-Smith, A.C., Pedersen, R.A., 2005b.Epigenetic status of human embryonic stem cells. Nat. Genet.37, 585–587.

Rugg-Gunn, P.J., Ferguson-Smith, A.C., Pedersen, R.A., 2007. Statusof genomic imprinting in human embryonic stem cells as revealedby a large cohort of independently derived and maintained lines.Hum. Mol. Genet. 16 (Spec No. 2), R243–R251.

Schatten, G., Mitalipov, S., 2009. Developmental biology: transgenicprimate offspring. Nature 459, 515–516.

Schramm, R.D., Paprocki, A.M., 2004. Strategies for the productionof genetically identical monkeys by embryo splitting. Reprod.Biol. Endocrinol. 2, 38.

Scott, C.T., 2006. Chimeras in the crosshairs. Nat. Biotechnol. 24,487–490.

Shapiro, E.M., Skrtic, S., Sharer, K., Hill, J.M., Dunbar, C.E.,Koretsky, A.P., 2004. MRI detection of single particles for cellularimaging. Proc. Natl. Acad. Sci. U S A 101, 10901–10906.

Simerly, C., Schatten, G., 1993. Techniques for localization ofspecific molecules in oocytes and embryos. Methods Enzymol.225, 516–553.

Simerly, C.R., Navara, C.S., Castro, C.A., Turpin, J.C., Redinger, C.J.,Mich-Basso, J.D., Jacoby, E.S., Grund, K.J., McFarland, D.A.,Oliver, S.L., Ben-Yehudah, A., Carlisle, D.L., Frost, P., Penedo, C.,Hewitson, L., Schatten, G., 2009. Establishment and characteri-zation of baboon embryonic stem cell lines: an Old World Primatemodel for regeneration and transplantation research. Stem CellRes. 2, 178–187.

Songsasen, N., Wildt, D.E., 2007. Oocyte biology and challenges indeveloping in vitro maturation systems in the domestic dog.Anim. Reprod. Sci. 98, 2–22.

St John, J., Lovell-Badge, R., 2007. Human–animal cytoplasmichybrid embryos, mitochondria, and an energetic debate. Nat.Cell Biol. 9, 988–992.

Stadtfeld, M., Brennand, K., Hochedlinger, K., 2008. Reprogrammingof pancreatic Beta cells into induced pluripotent stem cells. Curr.Biol. 18, 890–894.

Tachibana, M., Sparman, M., Sritanaudomchai, H., Ma, H., Clepper,L., Woodward, J., Li, Y., Ramsey, C., Kolotushkina, O.,Mitalipov, S., 2009. Mitochondrial gene replacement in primateoffspring and embryonic stem cells. Nature 461, 367–372.

Takada, T., Suzuki, Y., Kondo, Y., Kadota, N., Kobayashi, K., Nito,S., Kimura, H., Torii, R., 2002. Monkey embryonic stem cell linesexpressing green fluorescent protein. Cell Transplant. 11,631–635.

Takagi, Y., Takahashi, J., Saiki, H., Morizane, A., Hayashi, T., Kishi, Y.,Fukuda, H., Okamoto, Y., Koyanagi, M., Ideguchi, M., Hayashi, H.,Imazato, T., Kawasaki, H., Suemori, H., Omachi, S., Iida, H., Itoh,

N., Nakatsuji, N., Sasai, Y., Hashimoto, N., 2005. Dopaminergicneurons generated frommonkey embryonic stem cells function in aParkinson primate model. J. Clin. Invest. 115, 102–109.

Takahashi, J., 2006. Stem cell therapy for Parkinson's disease. ErnstSchering Res. Found. Workshop 229–244.

Takahashi, K., Ichisaka, T., Yamanaka, S., 2006. Identification ofgenes involved in tumor-like properties of embryonic stem cells.Methods Mol. Biol. 329, 449–458.

Tesar, P.J., Chenoweth, J.G., Brook, F.A., Davies, T.J., Evans, E.P.,Mack, D.L., Gardner, R.L., McKay, R.D., 2007. New cell lines frommouse epiblast share defining features with human embryonicstem cells. Nature 448, 196–199.

Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A.,Swiergiel, J.J., Marshall, V.S., Jones, J.M., 1998. Embryonicstem cell lines derived from human blastocysts. Science 282,1145–1147.

Trounson, A., 2006. The production and directed differentiation ofhuman embryonic stem cells. Endocr. Rev. 27, 208–219.

Vaca, P., Martin, F., Vegara-Meseguer, J.M., Rovira, J.M., Berna, G.,Soria, B., 2006. Induction of differentiation of embryonic stemcells into insulin-secreting cells by fetal soluble factors. StemCells 24, 258–265.

Warwick, B.L., Berry, R.O., 1949. Inter-generic and intra-specificembryo transfers in sheep and goats. J. Hered. 40, 297–303illust.

Wernig, M., Meissner, A., Foreman, R., Brambrink, T., Ku, M.,Hochedlinger, K., Bernstein, B.E., Jaenisch, R., 2007. In vitroreprogramming of fibroblasts into a pluripotent ES-cell-likestate. Nature 448, 318–324.

Wernig, M., Meissner, A., Cassady, J.P., Jaenisch, R., 2008. c-Myc isdispensable for direct reprogramming of mouse fibroblasts. CellStem Cell 2, 10–12.

Williams, T.J., Munro, R.K., Shelton, J.N., 1990. Production ofinterspecies chimeric calves by aggregation of Bos indicus andBos taurus demi-embryos. Reprod. Fertil. Dev. 2, 385–394.

Wood, S.A., Allen, N.D., Rossant, J., Auerbach, A., Nagy, A., 1993.Non-injection methods for the production of embryonic stemcell-embryo chimaeras. Nature 365, 87–89.

Xu, Y., Zhu, X., Hahm, H.S., Wei, W., Hao, E., Hayek, A., Ding, S.,2010. Revealing a core signaling regulatory mechanism forpluripotent stem cell survival and self-renewal by smallmolecules. Proc. Natl. Acad. Sci. U S A 107, 8129–8134.

Yamanaka, S., 2008. Induction of pluripotent stem cells from mousefibroblasts by four transcription factors. Cell Prolif. 41 (Suppl 1),51–56.

Yang, X.Y., Zhao, J.G., Li, H., Liu, H.F., Huang, Y., Huang, S.Z.,Zeng, F., Zeng, Y.T., 2008. Effect of individual heifer oocytedonors on cloned embryo development in vitro. Anim. Reprod.Sci. 104, 28–37.

Zhang, Y., Li, J., Villemoes, K., Pedersen, A.M., Purup, S., Vajta, G.,2007. An epigenetic modifier results in improved in vitroblastocyst production after somatic cell nuclear transfer. CloningStem Cells 9, 357–363.


Recommended