Open access

Human Testis–Derived Pluripotent Cells and Induced Pluripotent Stem Cells

Written By

Hideyuki Kobayashi, Koichi Nagao and Koichi Nakajima

Submitted: 15 August 2012 Published: 28 August 2013

DOI: 10.5772/55570

From the Edited Volume

Pluripotent Stem Cells

Edited by Deepa Bhartiya and Nibedita Lenka

Chapter metrics overview

2,319 Chapter Downloads

View Full Metrics

1. Introduction

Pregnancy rates achieved by intercourse in normal human couples are 20-25% per month, 75% by six months, and 90% by one year [1]. However, 15% of couples of unknown fertility status are unable to conceive a baby after one year of intercourse without contraception. For 30% of these couples, their infertility can be attributed to a male factor alone; in an additional 20%, failure to conceive is explained by the presence of both male and female factors [2,3,4]. Among couples known to be infertile, a male factor is involved in 50% of the cases. The most common causes of male infertility include abnormal sperm production or function, impaired delivery of sperm, and overexposure to certain gonadotoxins in the environment. The pathogenesis of male infertility can be attributed to a disorder of germ-cell proliferation and differentiation or to somatic cell dysfunction [5].

The induction of spermatogenesis depends on the complementary actions of FSH and testosterone. FSH establishes the requisite Sertoli cell population. In the prepubertal primate, FSH alone can induce proliferation of Sertoli cells and spermatogonia, but this does not result in qualitatively and quantitatively normal spermatogenesis unless testosterone is simultaneously present [6] [7]. Testosterone affects the functional completion of meiosis and post-meiotic sperm differentiation and maturation. LH stimulates Leydig cells to produce testosterone. Although FSH appears to play a more dominant role in the maintenance of primate spermatogenesis than in its initiation, normal spermatogenesis is best maintained by the combined effects of FSH and LH [6].

The most severe form of male infertility is nonobstructive azoospermia, which is typically characterized by small-volume testes and elevated FSH. Patients with this disorder cannot produce biological children. Although microdissection testicular sperm extraction (micro-TESE) is used to treat patients with nonobstructive azoospermia [8], this technique does not have a good success rate. Therefore, new approaches are needed to develop treatments for male infertility.

Stem cells have the potential to differentiate into a variety of functional cell types in the body, and their discovery has given rise to the fields of regenerative medicine and cloning. Stem cells are regulated by the particular microenvironment in which they reside; these microenvironments are referred to as niches. Male germline stem cells can continuously produce sperm throughout adulthood, and investigators have sought to develop methods using stem cells to improve or restore fertility.

Embryonic stem cells (ESCs) have the potential to differentiate into nearly every cell type in the body. As the cells differentiate, they lose the ability to develop into different tissues. In contrast, specific tissues (gastrointestinal, integumentary, spermatogenic, and hematopoietic systems) maintain their regenerative capacity in vivo, and in fact, stem cells have been functionally identified in a wide range of adult tissues. These adult stem cells are believed to hold great promise for tissue generation in clinical settings. Here, we provide a summary of the therapeutic potential of stem cells for the rejuvenation of fertility in infertile males. Our hope is that future research will provide a range of options for the preservation of male fertility or the reversal of infertility.

Advertisement

2. Differentiation and characterization of human primordial germ cells

Human primordial germ cells (PGCs) can be isolated from tissues and their identity confirmed by observing their migratory activity in vitro [9]. Cultured human PGCs become human embryonic germ cells (hEGCs) in vitro, in the presence of feeder cells, leukemia inhibitory factor (LIF), and basic fibroblast growth factor (bFGF) [10]. hEGCs express alkaline phosphatase (AP), OCT4, SOX2, NANOG, stage specific embryonic antigen (SSEA)-3, SSEA-4, TRA-1-60, and TRA-1-81, which are pluripotent stem cell markers. In vivo, human PGCs do not express FGF4, SOX2 [11] [12], TRA-1-60, or TRA-1-81 [13] [14], which are expressed by hESCs or hEGCs in vitro. The molecular signature of human PGCs in vivo can be characterized as C-KIT+, SOX2-, TRA1-60-, TRA1-81-, and FGF4-, in contrast with human pluripotent stem cell lines in vitro. (This information is summarized in Table 1.) However, the full complement of genes that are expressed specifically in human PGCs and their functions remain unclear.

Table 1.

Markers of human pluripotent stem cells and germ cells.

Advertisement

3. Spermatogonial stem cells

Spermatogenesis is a complex and tightly regulated process in which a small pool of germ-line stem cells ultimately gives rise to spermatozoa [15]. These stem cells, called spermatogonial stem cells (SSCs) are found in the basal compartment of the seminiferous epithelium, where they adhere to the basement membrane. SSC self-renewal ensures the maintenance of the stem cell pool, while their differentiation generates a large number of germ cells. Therefore, a balance between SSC self-renewal and differentiation in the adult testis is essential to maintain normal spermatogenesis and fertility throughout life. SSCs need to reside in a unique environment, or niche, that provides the factors necessary for their survival and potency. In mice, Sertoli cells in the testis are a crucial component of the spermatogonial stem cell niche. They produce glial cell line-derived neurotrophic factor (GDNF), a distant member of the TGFβ family, which controls SSC self-renewal [16]. Several groups have reported that adding GDNF to freshly isolated germ cells in culture results in the proliferation of SSCs [17,18]. Other factors within the niche influence the fate of SSCs. One example is colony-stimulating factor 1 (CSF1), which is produced by Leydig cells and some peritubular myoid cells [19], and plays a role in SSC self-renewal (Figure 1).

Figure 1.

Diagram of the spermatogonial stem cell (SSC) niche showing that extrinsic factors drive SSC maintenance and self-renewal. SSCs and Sertoli cells are attached to the basement membrane. Sertoli cells produce glial cell line-derived neurotrophic factor (GDNF) and basic fibroblast growth factor (bFGF). Leydig cells and peritubular cells produce colony-stimulating factor-1 (CSF-1).

The existence of SSCs was postulated almost 40 years ago on the basis of morphological studies [20] [21] [22] and observations of toxin-induced spermatogenic damage. The early studies of Clermont [23] [24] on human spermatogenesis revealed two types of spermatogonia, the Adark and Apale spermatogonia, which were differentiated by the staining pattern of their nucleus. Both cell types are generally considered stem cells [24,25]. Adark spermatogonia function as reverse stem cells that rarely divide, but can be triggered to self-renew in the case of injury or disease, while Apale spermatogonia are self-renewing stem cells [23,24,25,26]; they also divide into B spermatogonia, which further divide into spermatocytes [24].

In the last decade, molecular markers that can be used to identify and characterize human SSCs have been sought. A recent study reported that the expression of surface marker G protein coupled receptor 125 (GPR125) can be used in the isolation, characterization, and culture of putative human SSCs [27]. GPR125-positive spermatogonia are very rare, possibly limited to Adark spermatogonia or a sub-population of Apale spermatogonia. Human SSCs are also positive for some markers identified in mouse SSCs and other undifferentiated spermatogonia, including GFRA1, UCHL1 (PGP9.5), ZBTB16 (PLZF), and THY1 (CD90) [27,28]. We also have obtained evidence that THY1 is a potential surface marker for human SSCs [29].

Brinster and colleagues proved the existence of mouse SSCs by using unique approaches [30,31]. These investigators transplanted cells obtained as testicular homogenates expressing the LacZ gene into the seminiferous tubules of otherwise sterile mice with a Sertoli-cell-only pathology. After three months, the transplanted spermatogonial stem cells had engrafted and colonized the seminiferous tubules. Spermatogenesis was restored.

The clinical implications of this work are enormous. The findings suggest that the isolation, enrichment, and cryopreservation of spermatogonial stem cells prior to chemotherapy or radiation therapy, with later autologous transplantation, may offer the potential for the subsequent restoration of fertility. The development of this technique will be especially important for survivors of childhood cancer. Adult patients can also bank sperm for cryopreservation. However, most couples would prefer a naturally conceived child. Work has progressed in many laboratories to partially enrich the spermatogonial stem cells of species ranging from mice to primates. Today, many urologists bank a testicular biopsy from patients about to undergo chemotherapy, with the expectation that technology will advance rapidly over the next 10 years and allow transplantation in the future.

Advertisement

4. Pluripotency of human testis–derived ESC–like cells

Previous studies have demonstrated that neonatal and adult germline stem cells (GSCs) can be self-reprogrammed into ESC-like cells, called germline-derived pluripotent stem cells [32,33,34,35]. In addition, Conrad et al. [36] reported that pluripotent cells can be derived from human testis, which those authors called human adult GSCs (haGSCs). Other research groups subsequently claimed that ESC-like cells could be obtained from cultures of human testicular cells [37,38,39]. Conrad and colleagues compared the global gene expression profile of hESCs and haGSCs, and concluded that the populations presented a similar gene expression profile, and thus, that the haGSCs were pluripotent. However, Ko et al. claimed that the gene expression profile of haGSCs differed substantially from the pluripotent profile of hESCs, determined by a number of laboratories [40]. For example, the haGSCs did not express NANOG, and had low OCT4 and SOX2 levels, but showed high levels of the fibroblast markers SNA12 and ACTA2 [40]. Ko and colleagues therefore suggested that the haGSCs originated from fibroblast cells, rather than from pluripotent tissue. They concluded that haGSCs were very similar to a human testicular fibroblast cell line (hTFCs) [40]. Conrad and colleagues argued that microarray data sets cannot be compared unless they are processed in parallel in the same experiment, suggesting that the similarity between haGSCs and hTFCs was inconclusive. However, studies on microarray results generated by different laboratories [41,42,43] have shown that findings from microarray analyses are comparable across multiple laboratories [44], particularly when a common platform and set of procedures are used. These findings justify the utility of microarray repositories, such as the GEO database [45], not only as data warehouses but also as resources for comparative and combinatory analyses of microarray data from different laboratories. In conclusion, the global gene expression analysis of haGSCs demonstrated that these cells resembled fibroblast hTFCs more than pluripotent hESCs.

Advertisement

5. Induced pluripotent stem (iPS) cells

The year 2006 saw the first description of mouse induced pluripotent stem cells (miPSCs), which were generated by the retrovirus-mediated transduction of four transcription factors (OCT3/4, SOX2, KLF4, and C-MYC) into mouse fibroblasts [46]. Human somatic cells can be reprogrammed to become human iPSCs via the introduction of a small set of genes, either those encoding OCT3/4, SOX2 and KLF4, with or without the addition of C-MYC, or an alternate combination of OCT3/4, SOX2, LIN28, and NANOG [47,48,49,50,51,52,53,54,55]. Human iPSCs (hiPSCs) have remarkable similarity to hESCs in terms of their morphology, in vitro characteristics, proliferation rate, gene expression, and ability to differentiate into mesoderm, endoderm, and ectoderm, both in vitro and in vivo, in teratoma assays [56,57].

In our laboratory, we induced iPS cells from adult human testicular tissue by introducing four transcription factors, OCT4, SOX2, KLF4, and C-MYC, using lentiviral vectors [58]. We also generated ES-like cells from 293FT cells by using OCT4, SOX2, NANOG, and LIN28 [59]. Finally, we generated iPS cells derived from the human testicular tissue of individuals with Klinefelter syndrome (KS, also called 47, XXY) [60].

Advertisement

6. Germline differentiation from ESCs and iPSCs in humans

Recent studies indicate that mouse [61,62,63,64,65] and human [66,67] [50,68,69,70,71] ESCs can differentiate in vitro into oocyte- or sperm-like cells. In particular, Clark et al. first reported the spontaneous differentiation of germ cells in embryoid bodies derived from human ESCs [66]. Male germline cells express specific RNA and protein markers, such as VASA. In 2009, Park et al. demonstrated that PGC-like cells can be differentiated from human iPSCs [50]. Subsequent reports on male germline differentiation from stem cells have used one of three approaches: (1) specific culture conditions, (2) manipulation of gene expression, and (3) purification of germ cells.

Culture conditions supporting differentiation into germline cells. Bucay et al. observed that as hESCs differentiate into putative germline cells, they also produce Sertoli-like support cells [69]. In addition, co-cultures of hESCs and hiPSCs with human fetal gonadal stromal cells [50], mouse Sertoli cells [72], or mouse embryonic fibroblasts [67] resulted in the increased efficiency of germ cell-like differentiation. Co-culture systems are used to mimic a suitable microenvironment for the growing germ cells. For the differentiation of germline-like cells from hESCs and hiPSCs, cytokines and other cell-signaling molecules are often added to the cultures. For example, BMP4 and other BMPs are added to promote PGC-like differentiation from hESCs and hiPSCs [73,74,75]. In addition, retinoic acid has been used to stimulate meiosis [75] [76]. Panula and colleagues reported the differentiation of fetal- and adult-derived iPSCs into germ cells, and showed that ~5% of human iPSCs differentiated into PGCs following induction with BMPs [77].

Manipulation of gene expression. By manipulating gene expression, researchers can regulate the cell lineage decisions of differentiating pluripotent stem cells. Overexpression of DAZL and VASA promotes PGC formation in differentiating human ESCs and iPSCs [78]. In addition, Kee and colleagues (2009) reported that hESCs differentiate into germline cells that initiate meiosis and progress to form haploid germ cells. These authors indicated that the overexpression of members of the DAZ gene family, DAZ, DAZL, and BOULE, promoted the progression of PGCs to meiosis and the production of haploid cells, a process that is unique to germ cell development [71].

Purification of germline cells. The isolation and purification of germline cells from stem cell cultures (ESCs and iPSCs) can be performed efficiently when specific antibodies for germ cell surface markers are available. To purify PGC-like cells from differentiating human ESCs and iPSCs, cell sorting with specific antibodies for SSEA1 [68], SSEA1 and C-KIT [50] [79], or CXCR4 [69] has been effective. In particular, Eguizabal et al. (2011) published a straightforward protocol for germline cell purification that requires only three steps. First, human iPSCs and hESCs are allowed to differentiate for 3 weeks in a monolayer, in the absence of growth cytokines. Second, the cells are cultured for 3 weeks in the presence of retinoic acid. Finally, after these 6 weeks of differentiation, the cells are sorted for a specific combination of surface markers (CD49f++, CD9+, CD90-, and SSEA4-), and the isolated fraction is cultured in the presence of LIF, bFGF, Forskolin, and CYP26 inhibitor for 4 more weeks [76].

Advertisement

7. Germline differentiation from porcine iPSCs, non–human iPSCs

Despite their undoubted promise as sources of cells for tissue transplants, many roadblocks remain against using human ESCs clinically. Particularly troubling is the lack of tests for the efficacy of such therapies and the safety of transferring these cells in animals whose anatomy and physiology resemble those of humans better than mouse models do [80] [81] [82] [83] [84]. The pig is a potentially useful model in this regard, because of its similarities to humans in organ size, immunology, and whole animal physiology [85] [86] [87]. It was reported that porcine somatic cells can be reprogrammed to form piPSCs [88]. However, no reports on germline development from piPSCs have been published to date.

Advertisement

8. Conclusions

Research on stem cells has shown remarkable progress over the past 5 years. In particular, the development of human iPSCs has opened new avenues into the generation of an in vitro disease model of male infertility. However, improvements are still needed before stem cells can be used clinically. For the treatment and diagnosis of male infertility, future advances may enable spermatids to be differentiated from germline stem cells or iPS cells. In addition, by examining patient-specific iPSCs that are defective in their ability to generate germ cells and comparing their differentiation capacity with that of normal human ESCs and iPSCs, researchers can hope to uncover the nature of male infertility and to design new methods to reverse it.

Advertisement

Acknowledgments

This study was supported in part by a Grant-in-Aid for Young Scientists (B) of the Japan Society for the Promotion of Science (JSPS) and a Grant of the Strategic Research Foundation Grant-aided Project for Private schools at Heisei 23th from Ministry of Education, Culture, Sports, Science and Technology of Japan, 2011-2015.

References

  1. 1. SpiraAEpidemiology of human reproduction. Hum Reprod 198611115
  2. 2. MacLeod JHuman male infertility. Obstet Gynecol Surv 19712633551
  3. 3. MosherW. DReproductive impairments in the United States, 1965-1982. Demography 19852241530
  4. 4. SimmonsF. AHuman infertility. N Engl J Med 195625511406contd.
  5. 5. LueYErkkilaKLiuP. YMaKWangCHikimA. Set alFate of bone marrow stem cells transplanted into the testis: potential implication for men with testicular failure. Am J Pathol 2007170899908
  6. 6. NieschlagESimoniMGromollJWeinbauerG. FRole of FSH in the regulation of spermatogenesis: clinical aspects. Clin Endocrinol (Oxf) 19995113946
  7. 7. AllanC. MCouseJ. FSimanainenUSpalivieroJJimenezMRodriguezKet alEstradiol induction of spermatogenesis is mediated via an estrogen receptor-{alpha} mechanism involving neuroendocrine activation of follicle-stimulating hormone secretion. Endocrinology 2010151280010
  8. 8. SchlegelP. NNonobstructive azoospermia: a revolutionary surgical approach and results. Semin Reprod Med 20092716570
  9. 9. KuwanaTFujimotoTActive locomotion of human primordial germ cells in vitro. Anat Rec 1983205216
  10. 10. ShamblottM. JAxelmanJWangSBuggE. MLittlefieldJ. WDonovanP. Jet alDerivation of pluripotent stem cells from cultured human primordial germ cells. Proc Natl Acad Sci U S A 1998951372631
  11. 11. PerrettR. MTurnpennyLEckertJ. JOSheaMSonneS. BCameronIT et al. The early human germ cell lineage does not express SOX2 during in vivo development or upon in vitro culture. Biol Reprod 2008788528
  12. 12. De JongJStoopHGillisA. JVan GurpR. Jvan de Geijn GJ, Boer M et al. Differential expression of SOX17 and SOX2 in germ cells and stem cells has biological and clinical implications. J Pathol 20082152130
  13. 13. KerrC. LHillC. MBlumenthalP. DGearhartJ. DExpression of pluripotent stem cell markers in the human fetal testis. Stem Cells 20082641221
  14. 14. KerrC. LHillC. MBlumenthalP. DGearhartJ. DExpression of pluripotent stem cell markers in the human fetal ovary. Hum Reprod 20082358999
  15. 15. De RooijD. GRussellL. DAll you wanted to know about spermatogonia but were afraid to ask. J Androl 20002177698
  16. 16. MengXLindahlMHyvonenM. EParvinenMDe RooijD. GHessM. Wet alRegulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science 2000287148993
  17. 17. Kanatsu-shinoharaMOgonukiNInoueKMikiHOguraAToyokuniSet alLong-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod 2003696126
  18. 18. KubotaHAvarbockM. RBrinsterR. LGrowth factors essential for self-renewal and expansion of mouse spermatogonial stem cells. Proc Natl Acad Sci U S A 20041011648994
  19. 19. OatleyJ. MOatleyM. JAvarbockM. RTobiasJ. WBrinsterR. LColony stimulating factor 1 is an extrinsic stimulator of mouse spermatogonial stem cell self-renewal. Development 200913611919
  20. 20. HuckinsCThe spermatogonial stem cell population in adult rats. 3. Evidence for a long-cycling population. Cell Tissue Kinet 1971433549
  21. 21. HuckinsCThe spermatogonial stem cell population in adult rats. II. A radioautographic analysis of their cell cycle properties. Cell Tissue Kinet 1971431334
  22. 22. HuckinsCThe spermatogonial stem cell population in adult rats. I. Their morphology, proliferation and maturation. Anat Rec 197116953357
  23. 23. ClermontYThe cycle of the seminiferous epithelium in man. Am J Anat 19631123551
  24. 24. ClermontYRenewal of spermatogonia in man. Am J Anat 196611850924
  25. 25. ClermontYSpermatogenesis in man. A study of the spermatogonial population. Fertil Steril 19661770521
  26. 26. ClermontYKinetics of spermatogenesis in mammals: seminiferous epithelium cycle and spermatogonial renewal. Physiol Rev 197252198236
  27. 27. HeZKokkinakiMJiangJDobrinskiIDymMIsolation, characterization, and culture of human spermatogonia. Biol Reprod 20108236372
  28. 28. DymMKokkinakiMHeZSpermatogonial stem cells: mouse and human comparisons. Birth Defects Res C Embryo Today 2009872734
  29. 29. KobayashiHNagaoKNakajimaKMiuraKIshiiNThy-1+ cells isolated from adult human testicular tissues express human embryonic stem cell genes OCT3/4 and NANOG and may include spermatogonial stem cells. Reprod Med Biol 2009
  30. 30. BrinsterR. LAvarbockM. RGermline transmission of donor haplotype following spermatogonial transplantation. Proc Natl Acad Sci U S A 199491113037
  31. 31. BrinsterR. LZimmermannJ. WSpermatogenesis following male germ-cell transplantation. Proc Natl Acad Sci U S A 19949111298302
  32. 32. KoKTapiaNWuGKimJ. BBravoM. JSassePet alInduction of pluripotency in adult unipotent germline stem cells. Cell Stem Cell 200958796
  33. 33. Kanatsu-shinoharaMInoueKLeeJYoshimotoMOgonukiNMikiHet alGeneration of pluripotent stem cells from neonatal mouse testis. Cell 2004119100112
  34. 34. SeandelMJamesDShmelkovS. VFalciatoriIKimJChavalaSet alGeneration of functional multipotent adult stem cells from GPR125+ germline progenitors. Nature 200744934650
  35. 35. Kanatsu-shinoharaMLeeJInoueKOgonukiNMikiHToyokuniSet alPluripotency of a single spermatogonial stem cell in mice. Biol Reprod 2008786817
  36. 36. ConradSRenningerMHennenlotterJWiesnerTJustLBoninMet alGeneration of pluripotent stem cells from adult human testis. Nature 20084563449
  37. 37. GolestanehNKokkinakiMPantDJiangJDestefanoDFernandez-buenoCet alPluripotent stem cells derived from adult human testes. Stem Cells Dev 200918111526
  38. 38. KossackNMenesesJShefiSNguyenH. NChavezSNicholasCet alIsolation and characterization of pluripotent human spermatogonial stem cell-derived cells. Stem Cells 20092713849
  39. 39. MizrakS. CChikhovskayaJ. VSadri-ardekaniHVan DaalenSKorverC. MHovinghS. Eet alEmbryonic stem cell-like cells derived from adult human testis. Hum Reprod 20102515867
  40. 40. KoKArauzo-bravoM. JTapiaNKimJLinQBernemannCet alHuman adult germline stem cells in question. Nature 2010E1; discussion E3.
  41. 41. BammlerTBeyerR. PBhattacharyaSBoormanG. ABoylesABradfordB. Uet alStandardizing global gene expression analysis between laboratories and across platforms. Nat Methods 200523516
  42. 42. LarkinJ. EFrankB. CGavrasHSultanaRQuackenbushJIndependence and reproducibility across microarray platforms. Nat Methods 2005233744
  43. 43. IrizarryR. AWarrenDSpencerFKimI. FBiswalSFrankB. Cet alMultiple-laboratory comparison of microarray platforms. Nat Methods 2005234550
  44. 44. SherlockGOf fish and chips. Nat Methods 2005232930
  45. 45. EdgarRDomrachevMLashA. EGene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res 20023020710
  46. 46. TakahashiKYamanakaSInduction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 200612666376
  47. 47. OkitaKIchisakaTYamanakaSGeneration of germline-competent induced pluripotent stem cells. Nature 20074483137
  48. 48. YuJVodyanikM. ASmuga-ottoKAntosiewicz-bourgetJFraneJ. LTianSet alInduced pluripotent stem cell lines derived from human somatic cells. Science 2007318191720
  49. 49. WoltjenKMichaelI. PMohseniPDesaiRMileikovskyMHamalainenRet alpiggyBac transposition reprograms fibroblasts to induced pluripotent stem cells. Nature 200945876670
  50. 50. ParkT. SGalicZConwayA. ELindgrenAVan HandelB. JMagnussonMet alDerivation of primordial germ cells from human embryonic and induced pluripotent stem cells is significantly improved by coculture with human fetal gonadal cells. Stem Cells 20092778395
  51. 51. ZouJMaederM. LMaliPPruett-millerS. MThibodeau-begannySChouB. Ket alGene targeting of a disease-related gene in human induced pluripotent stem and embryonic stem cells. Cell Stem Cell 2009597110
  52. 52. NakagawaMKoyanagiMTanabeKTakahashiKIchisakaTAoiTet alGeneration of induced pluripotent stem cells without Myc from mouse and human fibroblasts. Nat Biotechnol 2008261016
  53. 53. TakahashiKOkitaKNakagawaMYamanakaSInduction of pluripotent stem cells from fibroblast cultures. Nat Protoc 2007230819
  54. 54. LiuHZhuFYongJZhangPHouPLiHet alGeneration of induced pluripotent stem cells from adult rhesus monkey fibroblasts. Cell Stem Cell 2008358790
  55. 55. TakahashiKTanabeKOhnukiMNaritaMIchisakaTTomodaKet alInduction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 200713186172
  56. 56. YamanakaSStrategies and new developments in the generation of patient-specific pluripotent stem cells. Cell Stem Cell 200713949
  57. 57. YamanakaSPluripotency and nuclear reprogramming. Philos Trans R Soc Lond B Biol Sci 2008363207987
  58. 58. KobayashiHNakajimaKOkaYTaiTNagaoKIshiiNReprogramming of adult human testicular cells by our transcription factors (OCT4, SOX2, KLF4, and C-MYC). Reprod Med Biol 201110105112
  59. 59. OkaYNakajimaKNagaoKMiuraKIshiiNKobayashiHFT cells transduced with four transcription actors (OCT4, SOX2, NANOG, and LIN28) generate aberrant ES-like cells. J. Stem cell and Regenerative Medicine 20103149156
  60. 60. KobayashiHPluripotent stem cells induced from testicular tissue with Klinefelter syndrome (47, XXY) by four ranscription factors (OCT4, SOX2, KLF4, and C-MYC). Methodological Advances in the Culture, Manipulation and Utilization of Embryonic Stem Cells for Basic and Practical Applications 2011295306
  61. 61. HubnerKFuhrmannGChristensonL. KKehlerJReinboldRDe La FuenteRet alDerivation of oocytes from mouse embryonic stem cells. Science 200330012516
  62. 62. ToyookaYTsunekawaNAkasuRNoceTEmbryonic stem cells can form germ cells in vitro. Proc Natl Acad Sci U S A 20031001145762
  63. 63. GeijsenNHoroschakMKimKGribnauJEgganKDaleyG. QDerivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 200442714854
  64. 64. QingTShiYQinHYeXWeiWLiuHet alInduction of oocyte-like cells from mouse embryonic stem cells by co-culture with ovarian granulosa cells. Differentiation 20077590211
  65. 65. NaganoM. CIn vitro gamete derivation from pluripotent stem cells: progress and perspective. Biol Reprod 20077654651
  66. 66. ClarkA. TBodnarM. SFoxMRodriquezR. TAbeytaM. JFirpoM. Tet alSpontaneous differentiation of germ cells from human embryonic stem cells in vitro. Hum Mol Genet 20041372739
  67. 67. WestF. DMachacekD. WBoydN. LPandiyanKRobbinsK. RSticeS. LEnrichment and differentiation of human germ-like cells mediated by feeder cells and basic fibroblast growth factor signaling. Stem Cells 200826276876
  68. 68. TilgnerKAtkinsonS. PGolebiewskaAStojkovicMLakoMArmstrongLIsolation of primordial germ cells from differentiating human embryonic stem cells. Stem Cells 200826307585
  69. 69. BucayNYebraMCirulliVAfrikanovaIKaidoTHayekAet alA novel approach for the derivation of putative primordial germ cells and sertoli cells from human embryonic stem cells. Stem Cells 2009276877
  70. 70. TilgnerKAtkinsonS. PYungSGolebiewskaAStojkovicMMorenoRet alExpression of GFP under the control of the RNA helicase VASA permits fluorescence-activated cell sorting isolation of human primordial germ cells. Stem Cells 2010288492
  71. 71. KeeKAngelesV. TFloresMNguyenH. NReijo Pera RA. Human DAZL, DAZ and BOULE genes modulate primordial germ-cell and haploid gamete formation. Nature 20094622225
  72. 72. GeensMSermonK. DVan de Velde H, Tournaye H. Sertoli cell-conditioned medium induces germ cell differentiation in human embryonic stem cells. J Assist Reprod Genet 20112847180
  73. 73. WestF. DRoche-riosM. IAbrahamSRaoR. RNatrajanM. SBacanamwoMet alKIT ligand and bone morphogenetic protein signaling enhances human embryonic stem cell to germ-like cell differentiation. Hum Reprod 20102516878
  74. 74. KeeKGonsalvesJ. MClarkA. TPeraR. ABone morphogenetic proteins induce germ cell differentiation from human embryonic stem cells. Stem Cells Dev 2006158317
  75. 75. RichardsMFongC. YBongsoAComparative evaluation of different in vitro systems that stimulate germ cell differentiation in human embryonic stem cells. Fertil Steril 20109398694
  76. 76. EguizabalCMontserratNVassenaRBarraganMGarretaEGarcia-quevedoLet alComplete meiosis from human induced pluripotent stem cells. Stem Cells 201129118695
  77. 77. PanulaSMedranoJ. VKeeKBergstromRNguyenH. NByersBet alHuman germ cell differentiation from fetal- and adult-derived induced pluripotent stem cells. Hum Mol Genet 20112075262
  78. 78. MedranoJ. VRamathalCNguyenH. NSimonCReijo Pera RA. Divergent RNA-binding proteins, DAZL and VASA, induce meiotic progression in human germ cells derived in vitro. Stem Cells 20123044151
  79. 79. AflatoonianBRubanLJonesMAflatoonianRFazeliAMooreH. DIn vitro post-meiotic germ cell development from human embryonic stem cells. Hum Reprod 20092431509
  80. 80. VackovaIUngrovaALopesFPutative embryonic stem cell lines from pig embryos. J Reprod Dev 200753113749
  81. 81. KeeferC. LPantDBlombergLTalbotN. CChallenges and prospects for the establishment of embryonic stem cell lines of domesticated ungulates. Anim Reprod Sci 20079814768
  82. 82. BreviniT. AAntoniniSCilloFCrestanMGandolfiFPorcine embryonic stem cells: Facts, challenges and hopes. Theriogenology 2007Suppl 1:S20613
  83. 83. TalbotN. CBlomberg Le A. The pursuit of ES cell lines of domesticated ungulates. Stem Cell Rev 2008423554
  84. 84. HallVPorcine embryonic stem cells: a possible source for cell replacement therapy. Stem Cell Rev 2008427582
  85. 85. PratherR. SHawleyR. JCarterD. BLaiLGreensteinJ. LTransgenic swine for biomedicine and agriculture. Theriogenology 20035911523
  86. 86. BrandlUMichelSErhardtMBrennerPBurdorfLJockleHet alTransgenic animals in experimental xenotransplantation models: orthotopic heart transplantation in the pig-to-baboon model. Transplant Proc 2007395778
  87. 87. PiedrahitaJ. AMirBCloning and transgenesis in mammals: implications for xenotransplantation. Am J Transplant 2004Suppl 64350
  88. 88. EzashiTTeluguB. PAlexenkoA. PSachdevSSinhaSRobertsR. MDerivation of induced pluripotent stem cells from pig somatic cells. Proc Natl Acad Sci U S A 2009106109938

Written By

Hideyuki Kobayashi, Koichi Nagao and Koichi Nakajima

Submitted: 15 August 2012 Published: 28 August 2013