5
Livestock Induced Pluripotent Stem Cells Y Lu 1,2 , JL Mumaw 1 , FD West 1 and SL Stice 1 1 Department of Animal and Dairy Science, Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; 2 State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China Contents Chimeric animals generated from livestock-induced pluripo- tent stem cells (iPSCs) have opened the door of opportunity to genetically manipulate species for the production of biomed- ical models, improving traits of agricultural importance and potentially providing a system to test novel iPSC therapies. The potential of pluripotent stem cells in livestock has long been recognized, with many attempts being chronicled to isolate, culture and characterize pluripotent cells from embryos. However, in most cases, livestock stem cells derived from embryonic sources have failed to reach a pluripotent state marked by the inability to form chimeric animals. The in-depth understanding of core pluripotency factors and the realization of how these factors can be harnessed to reprogram adult cells into an induced pluripotent state has changed the paradigm of livestock stem cells. In this review, we will examine the advancements in iPSC technology in mammalian and avian livestock species. Introduction The discovery of mouse embryonic stem cells (mESCs) in combination with new mammalian gene-altering technologies (Slightom et al. 1980; Folger et al. 1982) led to the first gene knockout mice (Koller et al. 1989; Thompson et al. 1989; Zijlstra et al. 1989). These advances in biomedical research allowed for the forma- tion of mice which modelled complex diseases generat- ing groundbreaking discoveries in the role of the genetics behind these diseases. Genetically engineered mice have come to be indispensible models for finding novel therapeutics and drug treatments, but they are often unsuitable to accurately model human diseases. The isolation and culture of pluripotent stem cells, cells capable of forming any cell type of the body, from embryos remained an elusive feat until 1981 when two groups successfully developed a co-culture system that was capable of maintaining mESCs in a pluripotent state with feeder cells secreting necessary factors to maintain pluripotency (Evans and Kaufman 1981; Martin 1981). These first mESC lines demonstrated characteristics that have come to define ESCs in all species. ESCs can be infinitely expanded, differentiated into all three germ layers (endoderm, ectoderm and mesoderm) in vitro as embryoid bodies (EBs) and in vivo in the form of teratomas (Evans and Kaufman 1981; Martin 1981), contributed to all tissue types including the germline in chimeric animals (Bradley et al. 1984) and acted as the inner cell mass (ICM) in tetraploid complementation assays (Eggan et al. 2001). The ability of ESCs to be continually expanded and contribute to the formation of the embryo proper are critical charac- teristics that enable complex genetic manipulations in producing animals for biomedical and agricultural purposes. However, producing ESCs capable of contributing to the germline has proven to be challenging in most species. Limitations in producing chimeras from ESCs have led to determination of two types of ESCs: naı¨ ve and primed (Ying et al. 2008). Naı¨ve (LIF dependent) mESCs possess a high propensity to form chimeric animals, have a high- level clongenicity and demonstrate key differences in epigenetics and gene expression from primed (FGF dependent) mESCs (Nichols and Smith 2009). Primed mESCs are similar in phenotype and cell signalling regulation to FGF-dependent hESC lines, which are driving the desire to derive naı¨ve ESCs in many species including humans (Lengner et al. 2010). The hypothesis is that naı¨ve state cells will generate a higher proportion of germline-competent pluripotent cells (Fig. 1). Isolation of Embryonic Stem Cells from Non-Primate and Rodent Species The development of pluripotent cultures have proven to be challenging outside of primates and rodents. While there has been progression in multiple species such as sheep (Sanna et al. 2010; Bao et al. 2011), swine (Strojek et al. 1990; Vassiliev et al. 2010), cattle (Stice et al. 1996; Cibelli et al. 1998; Saito et al. 2003), horses (Saito et al. 2002, 2006; Li et al. 2006), dog (Hatoya et al. 2006; Hayes et al. 2008), cats (Yu et al. 2008; Gomez et al. 2010), rabbits (Schoonjans et al. 1996; Wang et al. 2007), hamsters (Doetschman et al. 1988), mink (Sukoyan et al. 1993) and chickens (Pain et al. 1996; Horiuchi et al. 2004; van de Lavoir et al. 2006a,b), they all fail to meet the hallmarks of mESCs. Cells isolated from the ICM of various species often lack evidence of chimeric formation germline transmission, which limits their use in creating biomedical models and generating xenotherapeutic tissues. Many factors inhibit the devel- opment of chimeric-competent stem cell from non- primate and rodent species, including isolation and culture techniques. It is hoped that further understand- ing of pluripotency may aid in the development of chimeric-competent ESCs from these elusive species. Induced Pluripotent Stem Cells Core Transcriptional Factors Through transcriptional profiling, factors involved in the maintenance of pluripotency have been elucidated with Lin28, Pou5f1, Sox 2, Nanog and Nodal being actively transcribed in all ESCs and additionally Klf2 4 5 and C-myc being important in mESC (Cai et al. 2010). Nanog, Sox 2 and Pou5f1 have been Reprod Dom Anim 47 (Suppl. 4), 72–76 (2012); doi: 10.1111/j.1439-0531.2012.02057.x ISSN 0936-6768 Ó 2012 Blackwell Verlag GmbH

Livestock Induced Pluripotent Stem Cells

  • Upload
    y-lu

  • View
    213

  • Download
    1

Embed Size (px)

Citation preview

Livestock Induced Pluripotent Stem Cells

Y Lu1,2, JL Mumaw1, FD West1 and SL Stice1

1Department of Animal and Dairy Science, Regenerative Bioscience Center, University of Georgia, Athens, GA, USA; 2State Key Laboratory forConservation and Utilization of Subtropical Agro-Bioresources, Animal Reproduction Institute, Guangxi University, Nanning, Guangxi, China

Contents

Chimeric animals generated from livestock-induced pluripo-tent stem cells (iPSCs) have opened the door of opportunity togenetically manipulate species for the production of biomed-ical models, improving traits of agricultural importance andpotentially providing a system to test novel iPSC therapies.The potential of pluripotent stem cells in livestock has longbeen recognized, with many attempts being chronicled toisolate, culture and characterize pluripotent cells from embryos.However, in most cases, livestock stem cells derived fromembryonic sources have failed to reach a pluripotent statemarked by the inability to form chimeric animals. The in-depthunderstanding of core pluripotency factors and the realizationof how these factors can be harnessed to reprogram adult cellsinto an induced pluripotent state has changed the paradigm oflivestock stem cells. In this review, we will examine theadvancements in iPSC technology in mammalian and avianlivestock species.

Introduction

The discovery of mouse embryonic stem cells (mESCs)in combination with new mammalian gene-alteringtechnologies (Slightom et al. 1980; Folger et al. 1982)led to the first gene knockout mice (Koller et al. 1989;Thompson et al. 1989; Zijlstra et al. 1989). Theseadvances in biomedical research allowed for the forma-tion of mice which modelled complex diseases generat-ing groundbreaking discoveries in the role of thegenetics behind these diseases. Genetically engineeredmice have come to be indispensible models for findingnovel therapeutics and drug treatments, but they areoften unsuitable to accurately model human diseases.

The isolation and culture of pluripotent stem cells,cells capable of forming any cell type of the body, fromembryos remained an elusive feat until 1981 when twogroups successfully developed a co-culture system thatwas capable of maintaining mESCs in a pluripotentstate with feeder cells secreting necessary factors tomaintain pluripotency (Evans and Kaufman 1981;Martin 1981). These first mESC lines demonstratedcharacteristics that have come to define ESCs in allspecies. ESCs can be infinitely expanded, differentiatedinto all three germ layers (endoderm, ectoderm andmesoderm) in vitro as embryoid bodies (EBs) and in vivoin the form of teratomas (Evans and Kaufman 1981;Martin 1981), contributed to all tissue types includingthe germline in chimeric animals (Bradley et al. 1984)and acted as the inner cell mass (ICM) in tetraploidcomplementation assays (Eggan et al. 2001). The abilityof ESCs to be continually expanded and contribute tothe formation of the embryo proper are critical charac-teristics that enable complex genetic manipulations inproducing animals for biomedical and agricultural purposes.

However, producing ESCs capable of contributing tothe germline has proven to be challenging in mostspecies.

Limitations in producing chimeras from ESCs have ledto determination of two types of ESCs: naı̈ve and primed(Ying et al. 2008). Naı̈ve (LIF dependent)mESCs possessa high propensity to form chimeric animals, have a high-level clongenicity and demonstrate key differences inepigenetics and gene expression from primed (FGFdependent) mESCs (Nichols and Smith 2009). PrimedmESCs are similar in phenotype and cell signallingregulation to FGF-dependent hESC lines, which aredriving the desire to derive naı̈ve ESCs in many speciesincluding humans (Lengner et al. 2010). The hypothesis isthat naı̈ve state cells will generate a higher proportion ofgermline-competent pluripotent cells (Fig. 1).

Isolation of Embryonic Stem Cells fromNon-Primate and Rodent Species

The development of pluripotent cultures have proven tobe challenging outside of primates and rodents. Whilethere has been progression in multiple species such assheep (Sanna et al. 2010; Bao et al. 2011), swine (Strojeket al. 1990; Vassiliev et al. 2010), cattle (Stice et al. 1996;Cibelli et al. 1998; Saito et al. 2003), horses (Saito et al.2002, 2006; Li et al. 2006), dog (Hatoya et al. 2006;Hayes et al. 2008), cats (Yu et al. 2008; Gomez et al.2010), rabbits (Schoonjans et al. 1996; Wang et al.2007), hamsters (Doetschman et al. 1988), mink(Sukoyan et al. 1993) and chickens (Pain et al. 1996;Horiuchi et al. 2004; van de Lavoir et al. 2006a,b), theyall fail to meet the hallmarks of mESCs. Cells isolatedfrom the ICM of various species often lack evidence ofchimeric formation ⁄germline transmission, which limitstheir use in creating biomedical models and generatingxenotherapeutic tissues. Many factors inhibit the devel-opment of chimeric-competent stem cell from non-primate and rodent species, including isolation andculture techniques. It is hoped that further understand-ing of pluripotency may aid in the development ofchimeric-competent ESCs from these elusive species.

Induced Pluripotent Stem Cells CoreTranscriptional Factors

Through transcriptional profiling, factors involved inthe maintenance of pluripotency have been elucidatedwith Lin28, Pou5f1, Sox 2, Nanog and Nodal beingactively transcribed in all ESCs and additionallyKlf2 ⁄4 ⁄5 and C-myc being important in mESC(Cai et al. 2010). Nanog, Sox 2 and Pou5f1 have been

Reprod Dom Anim 47 (Suppl. 4), 72–76 (2012); doi: 10.1111/j.1439-0531.2012.02057.x

ISSN 0936-6768

� 2012 Blackwell Verlag GmbH

identified as ‘core’ factors, demonstrating both regionaland temporal expression in pluripotent cells of the ICM(Nichols et al. 1998; Avilion et al. 2003; Hart et al.2004). These ‘core’ factors are kept in a careful balancethrough positive autoregulation and synergistic regula-tion of other critical pluripotency factors (Boyer et al.2005). Additionally, Nanog has been identified as thekey factor regulating the establishment of the pluripo-tent epigenome (Silva et al. 2006) (Fig. 1). Understand-ing of the role and existence of these core pluripotencyfactors was pivotal in developing cellular reprogram-ming via the overexpression of transcription factors.

Generation of a pluripotent state was only possible inthe case of cell fusion and somatic cell nuclear transfer(McLaren 2000; Do and Scholer 2004) until discovery ofinduced pluripotency by Yamanaka (Takahashi andYamanaka 2006). Starting with 24 prospective genesknown to play a role in pluripotency maintenance, fourfactors (Pou5f1, Sox2, Klf4 and C-myc) were capable ofinitiating an ESC-like state when transduced intosomatic cells (Takahashi and Yamanaka 2006). Usingthe same approach in human fibroblasts, POU5F1,SOX2, KLF4 and C-MYC (Takahashi et al. 2007), orSOX2, POU5F1, NANOG and LIN28 (Yu et al. 2007)also produced iPSCs. Further, experimentation withcellular reprogramming has demonstrated that the onlyindispensible factors are POU5F1 and SOX2 (Eminliet al. 2008; Huangfu et al. 2008; Kim et al. 2008; Parket al. 2008), but the use of NANOG, LIN28, C-MYCand KLF-4 greatly increased reprogramming efficiency.

Although Nanog has been found to be dispensable forthe initiation of reprogramming, it is indispensible for apluripotent state and in its absence, cells can only bepartially reprogrammed (Takahashi and Yamanaka2006; Sridharan et al. 2009). Selecting iPSCs usingNanog expression has enabled more efficient chimerismand increases the incident of germline transmission(Okita et al. 2007). This requirement for Nanog furtherunderscores this gene as a ‘core’ member of pluripotencyfactors, being a gateway to the pluripotent ground state(Silva et al. 2009; Theunissen et al. 2011).

Induced Pluripotent Stem Cell in LivestockSpecies

Recent advances in iPSCs may overcome the roadblocksto establishing pluripotent lines in previously unobtain-able species and demonstrate that the factors of pluri-

potency can cross phylogenic lines. We have shown thathuman factors can be used on pig mesenchymal stemcells (MSCs) to generate iPSCs capable of generatingchimeras with germline transmission (West et al. 2011).These same human pluripotent factors can also formiPSCs from avian species, demonstrating that thesefactors are evolutionarily conserved (Lu et al. 2012).The list of livestock species that show the minimal levelof pluripotency (in vitro and teratoma formation)continues to rapidly increase and now includes sheep(Bao et al. 2011; Li et al. 2011), pigs (Esteban et al.2009; Ezashi et al. 2009; Wu et al. 2009; West et al.2010), rabbits (Honda et al. 2010) and horses (Nagyet al. 2011). Although these discoveries provide hope forthe future with these species in regenerative medicine,most studies have either not tested or reported successfulchimerism. In the pig and quail, we have generatedchimeras using iPSCs reprogrammed using all sixreprogramming factors [POU5F1, NANOG, LIN28,SOX2, KLF4 and C-MYC (PNLSKC)]; the system usedto generate these cells and characteristics is discussedbelow.

Porcine iPSC Generated Using Lentiviral-Based Overexpression of POU5F1, NANOG,LIN28, SOX2, KLF4 and C-MYC

In our porcine studies, we use the lentiviral humanreprogramming factors viPS� kit to overexpress thereprogramming factors PNLSKC driven by an EF1apromoter (West et al. 2010, 2011) to routinely establishmultiple independent stable porcine iPSC (piPSC) lines.These cells consistently demonstrate pluripotency char-acteristics with in vitro and in vivo tests. Our piPSCsdemonstrated, for the first time, that piPSC were capableof contributing to chimeric offspring (West et al. 2010,2011). After initial reprogramming of porcine MSCs,early emerging colonies grown on feeder cells did notappear to be fully reprogrammed; however, once piPSCswere transferred to feeder-free conditions, they showedexpression of the introduced human POU5F1 andSOX2 genes, and independent stem markers SSEA4and TRA 1-81. This proliferative and karyotypic stablecell line has been expanded for 100+ passages withoutchange in phenotype and continues to show robust invitro differentiation potential. Ultimately, injection ofpiPSCs into developing porcine embryos led to theproduction of eight foetuses and 29 live offspringpositive for the human POU5F1 (hPOU5F1) and ⁄orNANOG (hNANOG) genes, indicating that piPSCs hadsuccessfully integrated. Examination of foetal tissuesshowed high levels of chimerism with tissues from allthree germ layers including brain, skin, liver, pancreas,stomach, heart, kidney and spleen being positive for thehPOU5F1 gene. Interestingly, placenta tissue waspositive for hPOU5F1, suggesting that these cellscontributed to the extra-embryonic ectoderm. All spe-cies-specific iPSCs should be tested for ability tocontribution to chimeric offspring to be consideredtruly pluripotent, with the exception of humans, becauseof ethical and moral reasons. This validation is needed,given the reports that less pluripotent mouse iPSC(miPSC) lines can be generated, form teratomas,

Fig. 1. Nanog is the gateway to pluripotency (adopted and modifiedfrom Silva et al. 2009)

Livestock-Induced Pluripotent Stem Cells 73

� 2012 Blackwell Verlag GmbH

differentiate in vitro, but do not contribute to livechimeric offspring when injected into pre-implantationembryos (Nichols and Smith 2009). More recently, weconfirmed that piPSCs were capable of germline chime-rism for the first time in a non-rodent species (West et al.2010). We obtained a live birth of a transgenic piglet thatpossessed genome integration of the hPOU5F1 andhNANOG genes. Germline-competent piPSCs couldcompletely change the paradigm of how transgenicanimals are produced by making it possible to performmultiple gene knockouts and ⁄or knock-ins to producebiomedical pig models or improve economically impor-tant characteristics for production.

Perhaps of even more importance may be the potentialuse of piPSCs to test the efficacy and safety of noveliPSC therapies in allogeneic or autologous large animalpig models with similarities in anatomy and physiologyto humans. Since the advent of miPSCs, much of thediscussion around this technology has focused on usinga patient’s own cells to generate iPSCs for transplanta-tion. However, the scientific community has questionedthe predictive potential of rodent studies with respect tooutcomes in humans. Recently, derivation of photore-ceptors from piPSCs and the subsequent transplant ofthese cells into pigs devoid of rod photoreceptors whichresulted in successful engrafted and showed strong signsof integrating into the eye tissue has been accomplished(Zhou et al. 2011). This study highlights how the pigcould model potential stem cell therapies. Tumorigenic-ity of iPSCs is another critical concern as previousresearch in the mouse showed that iPSC-derived chime-ras possessed large numbers of tumours. In an initial testto determine the tumorigenicity of piPSCs, our researchgroup performed gross and histological examination ofchimeric animals derived from piPSCs at 2, 7 and 9 months,with all animals developing normally and absent oftumours, although tissue samples from the brain, skin,lung, pancreas, liver, heart and kidney were positive forhPOU5F1 DNA (West et al. 2011). The development ofgermline-competent piPSCs that do not producetumours presents an exciting and powerful translationalmodel to study the efficacy and safety of stem celltherapies and perhaps to efficiently produce complextransgenic animals.

Avian iPSC Generated by Overexpressionof Human Reprogramming Factors

Access to pluripotent stem cells in birds would be ofsignificance in developmental biology studies andtransgenic animal research. It has been demonstratedthat the mechanisms by which POU5F1 and NANOGregulate pluripotency and self-renewal are not exclu-sive to mammals (Lavial et al. 2007), indicating auniversal reprogramming process may exist; however,because of the lack of knowledge of reprogrammingfactors in phylogenetically diverse species, the use ofspecies-specific reprogramming factors to derive iPSCsin these animals is unlikely. In a recent study, wegenerated avian iPSCs using the viPS� kit followingthe established protocol from our piPSCs (West et al.2010). Quail embryonic fibroblasts were transducedwith reprogramming factors and formed quail iPSCs

(qiPSCs) that emerged as colonies in feeder co-cultureand eventually were maintained in feeder-free condi-tions. The derived qiPSC showed typical iPSC char-acteristics as seen in other species (Takahashi andYamanaka 2006; West et al. 2010): they displayedhigh nuclear-to-cytoplasm ratio, prominent nucleoliand telomerase activity >11 fold higher than theparent quail fibroblast cell line. qiPSC also stainedpositive for AP, PAS, POU5F1 and SOX2 and couldbe maintained for >50 passages without loss ofphenotype. In vitro differentiation of qiPSCs showedthat these cells were able to form all three germ layersin EBs and form quail–chick chimeras after injectioninto stage X chicken embryo, demonstrating the bonafide pluripotency of these cells (Lu et al. 2012). Thisfinding in qiPSCs is the first report of the widelyconserved nature of the cellular reprogramming pro-cess in birds, suggesting that it may be a universaltool to derive iPSCs from phylogenetically diversespecies.

As a model animal, avian species have a long historyof providing insight into aspects of developmentalbiology and disease. Avian species are relatively smallin size and the embryo can be easily accessed formanipulation enabling cells or tissues to be trans-planted, which is not possible in mammals (Kulesaet al. 2010). Previous publications reported the differ-entiation of chicken embryonic germ cells (EGs) intomultiple lineages, indicating that these cells may beutilized to perform transplantation studies in birds (Wuet al. 2010). Quail iPSCs are easily maintained in feeder-free and chemical-defined culture systems, and form EBsand demonstrate differentiation into all three germlayers: TUJ1 (ectoderm), SOX17 (endoderm) and alphasmooth muscle actin (aSMA, mesoderm). qiPSCs werealso found to be capable of directed neural differenti-ation expressing the neuron makers Hu C ⁄D and MAP2and the astrocyte and oligodendrocyte proteins GFAPand O4, respectively. These data demonstrated efficientspontaneous and directed differentiation of qiPSCs andthe potential opportunity for use in cell transplantationfor the purpose of developmental and regenerative therapyresearch.

Contribution of iPSCs to chimeric animals is con-sidered the most stringent criteria of pluripotency,indicating cells existing in a naı̈ve or ground state. Wehave recently shown that qiPSCs are capable ofcontributing to the live hatch of chicken–quail chime-ras, even after extended culture (Lu et al. 2012). Inprevious reports, chicken pluripotent cells have beenused to generate chimeric birds (van de Lavoir et al.2006a,b). However, maintaining pluripotent cells inprolonged culture has resulted in decreased chimerismand germline transmission (van de Lavoir et al.2006a,b). qiPSCs in our study are also capable ofclonal expansion after genetic modification, which hasnot been demonstrated before in pluripotent avian linesand would facilitate transgenic animal production.Techniques such as homologous recombinant offerunique opportunities to produce transgenic proteins ineggs, with important pharmaceutical use, or to studygene function in developmental processes using anavian model.

74 Y Lu, JL Mumaw, FD West and SL Stice

� 2012 Blackwell Verlag GmbH

Conclusion

Beyond expression, we believe the extent and durationof reprogramming factors present in the somatic cellsmay have been important in the generating the firstchimeric-competent livestock iPSCs. Not only is expres-sion needed, but expression levels may need to bedemonstrably higher to achieve a fully reprogrammedstate. Stromal tissue-sourced MSCs express some corepluripotent factors such as POU5F1, but at much lowerlevels than pluripotent stem cells. Bovine ESCs expressPOU5F1 protein, REX1 mRNA and SSEA-1 andSSEA-4 markers and are capable of forming EBs invitro and teratomas in vivo, but not chimeras. The samemarkers and genes including REX1 were expressed insheep iPSC and also have not shown chimerism ability(Bao et al. 2011). Although we have not quantifiedexpression levels of exogenous and endogenous corefactors in our chimera forming livestock iPSC, humaniPSC lines generated using the same reprogramming

system and MOI exhibited extremely high expression ofpluripotent markers. REX1 was approximately 1000fold increased in hiPSC lines than the donor fibroblastcells, and NANOG and POU5F1 were both over 5000fold higher. The key to chimerism in livestock speciesmay be achieving the proper ratio and level of allendogenous and exogenous core pluripotent transcrip-tion factors.

Conflicts of interest

S.L.S. is partially employed and has stock ownership in ArunABiomedical who in collaboration with Thermo Sceintific developed theviPS kit mentioned here.

Funding sources

Georgia Research Alliance and the Bill and Melinda Gates Founda-tions.

ReferencesAvilion AA, Nicolis SK, Pevny LH, Perez L,

Vivian N, Lovell-Badge R, 2003: Multi-potent cell lineages in early mouse devel-opment depend on SOX2 function. GenesDev 17, 126–140.

Bao L, He L, Chen J, Wu Z, Liao J, Rao L,Ren J, Li H, Zhu H, Qian L, Gu Y, Dai H,Xu X, Zhou J, Wang W, Cui C, Xiao L,2011: Reprogramming of ovine adult fi-broblasts to pluripotency via drug-induc-ible expression of defined factors. Cell Res21, 600–608.

Boyer LA, Lee TI, Cole MF, Johnstone SE,Levine SS, Zucker JP, Guenther MG,Kumar RM, Murray HL, Jenner RG,Gifford DK, Melton DA, Jaenisch R,Young RA, 2005: Core transcriptionalregulatory circuitry in human embryonicstem cells. Cell 122, 947–956.

Bradley A, Evans M, Kaufman MH,RobertsonE,1984:Formationofgerm-linechimaeras from embryo-derived teratocar-cinoma cell lines. Nature 309, 255–256.

Cai J, Xie D, Fan Z, Chipperfield H,Marden J, Wong WH, Zhong S, 2010:Modeling co-expression across species forcomplex traits: insights to the difference ofhuman and mouse embryonic stem cells.PLoS Comput Biol 6, e1000707.

Cibelli JB, Stice SL, Golueke PJ, Kane JJ,Jerry J, Blackwell C, Ponce de Leon FA,Robl JM, 1998: Transgenic bovine chi-meric offspring produced from somaticcell-derived stem-like cells. Nat Biotech-nol 16, 642–646.

Do JT, Scholer HR, 2004: Nuclei of embry-onic stem cells reprogram somatic cells.Stem Cells 22, 941–949.

Doetschman T, Williams P, Maeda N, 1988:Establishment of hamster blastocyst-derived embryonic stem (ES) cells. DevBiol 127, 224–227.

Eggan K, Akutsu H, Loring J, Jackson-Grusby L, Klemm M, Rideout WM 3rd,Yanagimachi R, Jaenisch R, 2001: Hybridvigor, fetal overgrowth, and viability ofmice derived by nuclear cloning andtetraploid embryo complementation. ProcNatl Acad Sci USA 98, 6209–6214.

Eminli S, Utikal J, Arnold K, Jaenisch R,Hochedlinger K, 2008: Reprogrammingof neural progenitor cells into inducedpluripotent stem cells in the absence ofexogenous Sox2 expression. Stem Cells26, 2467–2474.

Esteban MA, Xu J, Yang J, Peng M, Qin D,Li W, Jiang Z, Chen J, Deng K, ZhongM,Cai J, Lai L, Pei D, 2009: Generation ofinduced pluripotent stem cell lines fromTibetan miniature pig. J Biol Chem 284,

17634–17640.Evans MJ, Kaufman MH, 1981: Establish-

ment in culture of pluripotential cells frommouse embryos. Nature 292, 154–156.

EzashiT,TeluguBP,AlexenkoAP,SachdevS,Sinha S, Roberts RM, 2009: Derivationof induced pluripotent stem cells from pigsomatic cells. Proc Natl Acad Sci USA106, 10993–10998.

Folger KR, Wong EA, Wahl G, CapecchiMR, 1982: Patterns of integration of DNAmicroinjected into cultured mammaliancells: evidence for homologous recombina-tion between injected plasmid DNA mol-ecules. Mol Cell Biol 2, 1372–1387.

Gomez MC, Serrano MA, Pope CE, JenkinsJA, Biancardi MN, Lopez M, Dumas C,Galiguis J, Dresser BL, 2010: Derivationof cat embryonic stem-like cells from invitro-produced blastocysts on homolo-gous and heterologous feeder cells. The-riogenology 74, 498–515.

Hart AH, Hartley L, Ibrahim M, Robb L,2004: Identification, cloning and expres-sion analysis of the pluripotency promot-ing Nanog genes in mouse and human.Dev Dyn 230, 187–198.

Hatoya S, Torii R, Kondo Y, Okuno T,Kobayashi K, Wijewardana V, Kawate N,Tamada H, Sawada T, Kumagai D, Sugi-ura K, Inaba T, 2006: Isolation and char-acterization of embryonic stem-like cellsfrom canine blastocysts. Mol Reprod Dev73, 298–305.

Hayes B, Fagerlie SR, Ramakrishnan A,Baran S, Harkey M, Graf L, Bar M,Bendoraite A, Tewari M, Torok-Storb B,2008: Derivation, characterization, and invitro differentiation of canine embryonicstem cells. Stem Cells 26, 465–473.

Honda A, Hirose M, Hatori M, Matoba S,Miyoshi H, Inoue K, Ogura A, 2010:Generation of induced pluripotent stemcells in rabbits: potential experimentalmodels for human regenerative medicine.J Biol Chem 285, 31362–31369.

Horiuchi H, Tategaki A, Yamashita Y,Hisamatsu H, Ogawa M, Noguchi T,Aosasa M, Kawashima T, Akita S,Nishimichi N, Mitsui N, Furusawa S,Matsuda H, 2004: Chicken leukemiainhibitory factor maintains chickenembryonic stem cells in the undifferenti-ated state. J Biol Chem 279, 24514–24520.

Huangfu D, Osafune K, Maehr R, Guo W,Eijkelenboom A, Chen S, Muhlestein W,Melton DA, 2008: Induction of pluripo-tent stem cells from primary humanfibroblasts with only Oct4 and Sox2. NatBiotechnol 26, 1269–1275.

Kim JB, Zaehres H, Wu G, Gentile L, Ko K,Sebastiano V, Arauzo-BravoMJ, RuauD,Han DW, Zenke M, Scholer HR, 2008:Pluripotent stem cells induced from adultneural stem cells by reprogramming withtwo factors. Nature 454, 646–650.

Koller BH, Hagemann LJ, Doetschman T,Hagaman JR, Huang S, Williams PJ, FirstNL, Maeda N, Smithies O, 1989: Germ-line transmission of a planned alterationmade in a hypoxanthine phosphoribosyl-transferase gene by homologous recombi-nation in embryonic stem cells. Proc NatlAcad Sci USA 86, 8927–8931.

Kulesa PM, Bailey CM, Cooper C, FraserSE, 2010: In ovo live imaging of avianembryos. Cold Spring Harb Protoc 2010,

pdb prot5446.Lavial F, Acloque H, Bertocchini F, Macle-

od DJ, Boast S, Bachelard E, Montillet G,Thenot S, SangHM, Stern CD, Samarut J,Pain B, 2007: The Oct4 homologue PouVand Nanog regulate pluripotency in chick-en embryonic stem cells. Development 134,3549–3563.

van de Lavoir MC, Diamond JH, LeightonPA, Mather-Love C, Heyer BS, Brad-shaw R, Kerchner A, Hooi LT, GessaroTM, Swanberg SE, Delany ME, EtchesRJ, 2006a: Germline transmission of

Livestock-Induced Pluripotent Stem Cells 75

� 2012 Blackwell Verlag GmbH

genetically modified primordial germ cells.Nature 441, 766–769.

van de LavoirMC,Mather-Love C, LeightonP,DiamondJH,HeyerBS,RobertsR,ZhuL, Winters-Digiacinto P, Kerchner A,Gessaro T, Swanberg S, Delany ME,Etches RJ, 2006b: High-grade transgenicsomatic chimeras from chicken embryonicstem cells. Mech Dev 123, 31–41.

Lengner CJ, Gimelbrant AA, Erwin JA,Cheng AW, Guenther MG, WelsteadGG, Alagappan R, Frampton GM, XuP, Muffat J, Santagata S, Powers D,Barrett CB, Young RA, Lee JT, JaenischR, Mitalipova M, 2010: Derivation ofpre-X inactivation human embryonicstem cells under physiological oxygenconcentrations. Cell 141, 872–883.

LiX,ZhouSG,ImrehMP,Ahrlund-RichterL,Allen WR, 2006: Horse embryonic stemcell lines from the proliferation of inner cellmass cells. Stem Cells Dev 15, 523–531.

Li Y, Cang M, Lee AS, Zhang K, Liu D,2011: Reprogramming of sheep fibro-blasts into pluripotency under a drug-inducible expression of mouse-deriveddefined factors. PLoS ONE 6, e15947.

Lu Y, West FD, Jordan BJ, Mumaw JL,Jordan ET, Gallegos-Cardenas A, Beck-stead RB, Stice SL, 2012: Avian-inducedpluripotent stem cells derived using hu-man reprogramming factors. Stem CellsDev 21, 394–403.

Martin GR, 1981: Isolation of a pluripotentcell line from early mouse embryos cul-tured in medium conditioned by terato-carcinoma stem cells. Proc Natl Acad SciUSA 78, 7634–7638.

McLaren A, 2000: Cloning: pathways to apluripotent future. Science288,1775–1780.

Nagy K, Sung HK, Zhang P, Laflamme S,Vincent P, Agha-Mohammadi S, WoltjenK, Monetti C, Michael IP, Smith LC,Nagy A, 2011: Induced pluripotent stemcell lines derived from equine fibroblasts.Stem Cell Rev 7, 693–702.

Nichols J, Smith A, 2009: Naive and primedpluripotentstates.CellStemCell4,487–492.

Nichols J, Zevnik B, Anastassiadis K, NiwaH, Klewe-Nebenius D, Chambers I,Scholer H, Smith A, 1998: Formation ofpluripotent stem cells in the mammalianembryo depends on the POU transcrip-tion factor. Cell 95, 379–391.

Okita K, Ichisaka T, Yamanaka S, 2007:Generation of germline-competent in-duced pluripotent stem cells. Nature448, 313–317.

Pain B, Clark ME, Shen M, Nakazawa H,Sakurai M, Samarut J, Etches RJ, 1996:Long-term in vitro culture and charac-terisation of avian embryonic stem cellswith multiple morphogenetic potentiali-ties. Development 122, 2339–2348.

Park IH, Zhao R, West JA, Yabuuchi A,Huo H, Ince TA, Lerou PH, Lensch MW,Daley GQ, 2008: Reprogramming of hu-man somatic cells to pluripotency withdefined factors. Nature 451, 141–146.

Saito S, Ugai H, Sawai K, Yamamoto Y,Minamihashi A, Kurosaka K, KobayashiY, Murata T, Obata Y, Yokoyama K,2002: Isolation of embryonic stem-likecells from equine blastocysts and theirdifferentiation in vitro. FEBS Lett531, 389–396.

Saito S, Sawai K, Ugai H, Moriyasu S,Minamihashi A, Yamamoto Y, HirayamaH, Kageyama S, Pan J, Murata T,Kobayashi Y, Obata Y, Yokoyama KK,2003: Generation of cloned calves andtransgenic chimeric embryos from bovineembryonic stem-like cells. Biochem Bio-phys Res Commun 309, 104–113.

Saito S, Sawai K, Minamihashi A, Ugai H,Murata T, Yokoyama KK, 2006: Deriva-tion, maintenance, and induction of thedifferentiation in vitro of equine embryonicstem cells. Methods Mol Biol 329, 59–79.

Sanna D, Sanna A, Mara L, Pilichi S,Mastinu A, Chessa F, Pani L, Dattena M,2010: Oct4 expression in in-vitro-producedsheep blastocysts and embryonic-stem-likecells. Cell Biol Int 34, 53–60.

Schoonjans L, Albright GM, Li JL, CollenD, Moreadith RW, 1996: Pluripotentialrabbit embryonic stem (ES) cells arecapable of forming overt coat color chi-meras following injection into blastocysts.Mol Reprod Dev 45, 439–443.

Silva J, Chambers I, Pollard S, Smith A,2006: Nanog promotes transfer of pluri-potency after cell fusion. Nature 441,

997–1001.Silva J, Nichols J, Theunissen TW, Guo G,

van Oosten AL, Barrandon O, Wray J,Yamanaka S, Chambers I, Smith A, 2009:Nanog is the gateway to the pluripotentground state. Cell 138, 722–737.

Slightom JL, Blechl AE, Smithies O, 1980:Human fetal G gamma- and A gamma-globin genes: complete nucleotidesequences suggest that DNA can beexchanged between these duplicatedgenes. Cell 21, 627–638.

Sridharan R, Tchieu J, Mason MJ, Ya-chechko R, Kuoy E, Horvath S, Zhou Q,Plath K, 2009: Role of the murine repro-gramming factors in the induction ofpluripotency. Cell 136, 364–377.

Stice SL, Strelchenko NS, Keefer CL, Mat-thews L, 1996: Pluripotent bovine embry-onic cell lines direct embryonicdevelopment following nuclear transfer.Biol Reprod 54, 100–110.

Strojek RM, Reed MA, Hoover JL, WagnerTE, 1990: A method for cultivating mor-phologically undifferentiated embryonicstem cells from porcine blastocysts. The-riogenology 33, 901–913.

Sukoyan MA, Vatolin SY, Golubitsa AN,Zhelezova AI, Semenova LA, Serov OL,1993: Embryonic stem cells derived frommorulae, inner cell mass, and blastocystsof mink: comparisons of their pluripoten-cies. Mol Reprod Dev 36, 148–158.

Takahashi K, Yamanaka S, 2006: Inductionof pluripotent stem cells from mouseembryonic and adult fibroblast culturesby defined factors. Cell 126, 663–676.

TakahashiK,TanabeK,OhnukiM,NaritaM,Ichisaka T, Tomoda K, Yamanaka S,2007: Induction of pluripotent stem cellsfrom adult human fibroblasts by definedfactors. Cell 131, 861–872.

Theunissen TW, van Oosten AL, Castelo-Branco G, Hall J, Smith A, Silva JC,2011: Nanog overcomes reprogrammingbarriers and induces pluripotency in min-imal conditions. Curr Biol 21, 65–71.

Thompson S, Clarke AR, Pow AM, HooperML, Melton DW, 1989: Germ line trans-

mission and expression of a correctedHPRT gene produced by gene targeting inembryonic stem cells. Cell 56, 313–321.

Vassiliev I, Vassilieva S, Beebe LF,McIlfatrick SM, Harrison SJ, NottleMB, 2010: Development of culture con-ditions for the isolation of pluripotentporcine embryonal outgrowths from invitro produced and in vivo derivedembryos. J Reprod Dev 56, 546–551.

Wang S, Tang X, Niu Y, Chen H, Li B, Li T,Zhang X, Hu Z, Zhou Q, Ji W, 2007:Generation and characterization of rabbitembryonic stem cells. Stem Cells 25, 481–489.

West FD, Terlouw SL, Kwon DJ, MumawJL, Dhara SK, Hasneen K, DobrinskyJR, Stice SL, 2010: Porcine induced plu-ripotent stem cells produce chimeric off-spring. Stem Cells Dev 19, 1211–1220.

West FD, Uhl EW, Liu Y, Stowe H, Lu Y,Yu P, Gallegos-Cardenas A, Pratt SL,Stice SL, 2011: Brief report: chimeric pigsproduced from induced pluripotent stemcells demonstrate germline transmissionand no evidence of tumor formation inyoung pigs. Stem Cells 29, 1640–1643.

Wu Z, Chen J, Ren J, Bao L, Liao J, Cui C,Rao L, Li H, Gu Y, Dai H, Zhu H, TengX, Cheng L, Xiao L, 2009: Generation ofpig induced pluripotent stem cells with adrug-inducible system. J Mol Cell Biol1, 46–54.

Wu Y, Ge C, Zeng W, Zhang C, 2010:Induced multilineage differentiation ofchicken embryonic germ cells via embry-oid body formation. Stem Cells Dev 19,

195–202.YingQL,Wray J,Nichols J, Batlle-MoreraL,

Doble B, Woodgett J, Cohen P, Smith A,2008: The ground state of embryonic stemcell self-renewal. Nature 453, 519–523.

Yu J, Vodyanik MA, Smuga-Otto K, Anto-siewicz-Bourget J, Frane JL, Tian S, NieJ, Jonsdottir GA, Ruotti V, Stewart R,Slukvin II, Thomson JA, 2007: Inducedpluripotent stem cell lines derived fromhuman somatic cells. Science 318, 1917–1920.

Yu X, Jin G, Yin X, Cho S, Jeon J, Lee S,Kong I, 2008: Isolation and characteriza-tion of embryonic stem-like cells derivedfrom in vivo-produced cat blastocysts.Mol Reprod Dev 75, 1426–1432.

ZhouL,WangW,LiuY, deCastro JF,EzashiT, Telugu BP, Roberts RM, Kaplan HJ,DeanDC, 2011: Differentiation of inducedpluripotent stem cells of swine into rodphotoreceptors and their integration intothe retina. Stem Cells 29, 972–980.

Zijlstra M, Li E, Sajjadi F, Subramani S,Jaenisch R, 1989: Germ-line transmissionof a disrupted beta 2-microglobulin geneproduced by homologous recombinationin embryonic stem cells. Nature 342,

435–438.

Author’s address (for correspondence):YangqingLu, Department of Animal and DairyScience, Regenerative Bioscience Center,University of Georgia, 425 River Road,Athens, GA 30602, USA. E-mail: [email protected]

76 Y Lu, JL Mumaw, FD West and SL Stice

� 2012 Blackwell Verlag GmbH