当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2005年第2期 > 正文
编号:11340035
Immortalization of Mouse Germ Line Stem Cells
http://www.100md.com 《干细胞学杂志》
     a Department of Biology, University of Dayton, Dayton, Ohio, USA;

    b Department of Cell Biology, Georgetown University Medical Center, Washington, DC, USA

    Key Words. Type A spermatogonia ? SV40 ? Large T antigen ? Testis ? Germ line stem cell ? GFR-1 ? Glial cell line–derived neurotrophic factor (GDNF)

    Correspondence: Marie-Claude Hofmann, Ph.D., Department of Biology, University of Dayton, 300 College Park, Dayton, OH 45469–2320. Telephone: 937-229-2894; Fax: 937-229-2021; e-mail: Marie-Claude.Hofmann@notes.udayton.edu

    ABSTRACT

    Stem cells are unique cell populations that are able to undergo both self-renewal and differentiation and are found in the embryo, as well as in the adult animal. In the early mammalian embryo, pluripotent embryonic stem cells are derived from the blastocyst stage and have the ability to form any fully differentiated cell of the body. As the embryo develops, stem cells become restricted in their ability to form different lineages (multipotent stem cells). Multipotent stem cells are also found in a wide variety of adult tissues such as bone marrow and brain. However, in the adult animal, the ability of certain stem cells to differentiate can be restricted to only one cell lineage (unipotent stem cells). Examples of mammalian unipotent stem cells include the stem cells residing in the gut epithelium, the skin, and the seminiferous epithelium of the testis.

    A widely accepted model for spermatogonial development has been originally proposed by Huckins and Oakberg . In this scheme, the Asingle (As) spermatogonia are the putative stem cells. They can self-renew or differentiate into Apaired (Apr) spermatogonia that remain connected by an intercellular bridge. The Apr spermatogonia further divide to form chains of 4, 8, or 16 Aaligned (Aal) spermatogonia. Further, the Aal cells will differentiate into type A1 spermatogonia. The A1 spermatogonia resume division to form A2 to A4 spermatogonia. Next, A4 cells divide to form intermediate (In) spermatogonia, and In spermatogonia divide to produce type B spermatogonia. Finally, type B spermatogonia divide to form primary spermatocytes that will enter meiosis. Spermatogonial stem cells and their progeny are contained in the basal part of the germinal epithelium, in contact with the basement membrane, and they are in close association with the somatic Sertoli cells . Regulatory mechanisms mediated by growth factors produced by the Sertoli cells induce or inhibit the proliferation, differentiation, and further development of the germ cells .

    Spermatogonial stem cells can generate spermatogenesis when transplanted into the seminiferous tubules of an infertile male . Currently, there is a limited understanding of the molecular mechanisms that control the development of these cells into mature sperm. Spermatogonial stem cells exhibit a distinct phenotype such as the high expression of ?-1 and -6 integrins and specific light-scattering properties . While the stem cell identity of the As spermatogonia has not yet been rigorously demonstrated, their morphology and location in the seminiferous epithelium make them good candidates for being stem cells . As spermatogonia express GFR-1, the receptor for glial cell line–derived neurotrophic factor (GDNF) . As the As differentiate into Apr and Aal spermatogonia, they start expressing the surface receptor c-kit, which binds to stem cell growth factor (SCF) produced by Sertoli cells .

    The ability to isolate, culture, and manipulate the germ line stem cell in vitro would allow us to unravel the molecular mechanisms that drive the first steps of spermatogenesis and to characterize the signaling pathways that induce spermatogonial differentiation versus self-renewal. In turn, this could help us understand the origin of certain testicular neoplasias and the causes of male infertility. To look at these issues, an in vitro system in which these cells could be maintained in long-term cultures would be ideal. In the study reported here, we attempted to establish a mouse spermatogonial stem cell line using the large T antigen under the control of an inducible promoter. The resulting immortalized cells express detectable levels of protein and RNA specific for As and Apr spermatogonia such as the GFR-1 membrane receptor. Further analysis revealed the expression of additional markers accounting for a stem cell phenotype such as the genes oct-4, piwi12, and prame11 that have a role in stem cell maintenance and renewal in the germ line and other tissues.

    MATERIALS AND METHODS

    Cell Immortalization

    In our immortalization strategy, the ecdysone receptor from Drosophila was expressed from a vector called pVgRxR . The ecdysone-responsive promoter, which drives the expression of the large T-antigen gene, is on a second vector called pIND-LTAg. Both vectors were stably cotransfected into freshly isolated type A spermatogonia that were induced to express the LTAg when treated with Ponasterone A, an analog of ecdysone. While the cells escaped the hormonal control after a finite number of generations and expressed the LTAg constitutively, their growth remained slow, with a doubling time of about 3 days. After several rounds of subcloning by limiting dilution, a clone was obtained that we called C18-4. The cells appeared round to polygonal, with a round nucleus, and a large nucleus-to-cytoplasm ratio in phase contrast microscopy (Fig. 1).

    Figure 1. Morphology of the C18-4 cells in phase contrast microscopy. The cells are round to polygonal, with a round nucleus, and a large nucleus-to-cytoplasm ratio.

    Immunocytochemistry

    The purified C18-4 cells were stained by immunocytochemistry for the presence of the germ cell–specific Dazl protein using antisera nos. 149 and 150 . Figure 2 shows that Dazl is expressed in the nucleus when it is stained with antiserum no. 149 (Fig. 2A), and it is expressed in the nucleus and the cytoplasm when it is stained with antiserum no. 150 (Fig. 2C). The cells were also positive for GFR-1, the coreceptor of Ret, and a marker for germ line stem cells (Fig. 3A). Further, they stained positive for the Ret transmembrane receptor, a marker for spermatogonia and spermatocytes (Fig. 3C). The cell nuclei, in particular the nucleoli, stained positive for Oct-4, an early marker for the germ line (Fig. 3E). The expression of the c-kit receptor could not be revealed using immunocytochemistry, but it was detected by RT-PCR in early passages only (Fig. 4).

    Figure 2. C18-4 cells, immunostained for the Dazl RNA-binding protein. (A): Staining for Dazl, a general marker for germ cells, revealed by immunocytochemistry using antiserum no. 149 and horseradish peroxidase. (B): Negative control without primary antibody. (C): Staining for Dazl, a general marker for germ cells, revealed by immunocytochemistry using antiserum no. 150 and horseradish peroxidase. (D): Negative control without primary antibody.

    Figure 3. C18-4 cells, immunostained for GFR-1, the Ret transmembrane receptor, and the Oct-4 transcription factor. (A): Staining for GFR-1, revealed by immunocytochemistry and horseradish peroxidase (x100). (B): Negative control without primary antibody (x100). (C): Staining for Ret, revealed by immunocytochemistry and horseradish peroxidase (x100). (D): Negative control without primary antibody (x100). (E): Staining for Oct-4, revealed by immunocytochemistry and horseradish peroxidase (x40). (F): Negative control without primary antibody (x40).

    Figure 4. c-kit gene expression in the C18-4 cells visualized by reverse transcription polymerase chain reaction. Lane A: size markers; lane B: adult testis; lane D: C18-4 cell line; lane F: freshly isolated Sertoli cell. Lanes C, E, and G: negative controls without reverse transcriptase.

    Immunoprecipitation

    The expression of GFR-1 and Dazl in the C18-4 cell line was confirmed by immunoprecipitation (Fig. 5). To increase the specificity of detection, GFR-1 was immunoprecipitated using a polyclonal antibody made in goat, then revealed after blotting using a polyclonal antibody made in rabbit. In Figure 5A, lane E, we show that the C18-4 cell line produces a protein exhibiting a molecular weight of 42 kDA that corresponds to the molecular weight of GFR-1. As positive controls, we used protein extracts from developing brain, kidney, and testis (lanes B–D, respectively). As negative controls, we used NIH 3T3 cells and the Sertoli cell line 15P1 (lanes F and G) .

    Figure 5. Immunoprecipitation of GFR-1, Dazl, and Oct-4 using C18-4 protein extracts. (A): Immunoprecipitation of GFR-1. Lane A: molecular weight markers; lane B: adult kidney; lane C: developing brain (6-day-old); lane D: adult testis; lane E: C18-4 cell line; lane F: 15P1 Sertoli cell line; lane G: NIH 3T3 cell line. (B): Immunoprecipitation of Dazl. Lane A: molecular weight markers; lane B: adult testis; lane C: C18-4 cell line. (C): Immunoprecipitation of Oct-4. Lane A: C18-4 cell line; lane B: adult testis; lane C: molecular weight markers.

    The Dazl protein was immunoprecipitated and revealed after blotting using antiserum no. 149 . As seen in Figure 5B, a strong band with a molecular weight of approximately 33 kDa was revealed in the testis extract, corresponding to the molecular weight of Dazl . The same band was observed using C18-4 extracts (Fig. 5B). Similarly, the Oct-4 protein was revealed after immunoprecipitation (Fig. 5C).

    Finally, immunoprecipitation failed to detect the expression of GDNF in the purified cells, indicating that the C18-4 cells are not contaminated by Sertoli cells.

    Reverse Transcriptase Polymerase Chain Reaction

    Neither 3?HSD, a marker for Leydig cells, nor -actin, a marker for peritubular myoid cells, could be found by RT-PCR, further indicating that the cells are of germ cell origin. The absence of LDHC4 expression indicated that the cells are not spermatocytes.

    To further confirm the spermatogonial origin of the C18-4 cell line, we investigated by RT-PCR the expression of 36 spermatogonial-specific genes, which were previously identified by Wang and colleagues in the 6-day-old mouse testis. There was a RT-PCR product for the housekeeping gene Gapd in both cell lines and in the neonatal testis samples, indicating that the RNA isolated contained intact transcripts (data not shown). RT-PCR further revealed that all 36 spermatogonial-specific genes tested were expressed in the 6-day-old testis, as expected. None of these genes were expressed in the SF7 Sertoli cell line, confirming the specificity of the transcripts. The expression of 10 spermatogonial-specific genes was detected in the C18-4 cell line. The results of these 10 are reported in Table 1.

    Table 1. Spermatogonial genes expressed in the C18-4 cell line, arranged in alphabetical order; 6-day-old testes were used as positive control, and the Sertoli cell line SF7 as negative control

    One of the genes expressed by the C18-4 cells was the Ott gene (Fig. 6A), which is transcribed in vivo in the germ cells of testis and ovary . The primers used in this study can detect the presence of two bands by RT-PCR, corresponding to two isoforms, with sizes of 551 and 503 bp, respectively . As shown in Figure 6A, two bands of the correct size are expressed by the C18-4 cells (lane B). However, only one band (551 bp) was detectable in the neonatal testis sample (lane D). Sertoli cells do not express the Ott gene (lane F). Figure 6B shows that the C18-4 cell line also expresses Sycp1, a gene coding for a protein of the synaptonemal complex, which is already expressed in spermatogonia . The band obtained is of 311 bp, as expected (lane B). The same band is visible for the 6-day-old testis sample (lane D), but not in Sertoli cells (lane F). Figure 6C represents the expression of the gene Fth117 that codes for a testis-specific ferritin-binding protein . Fth117 is expressed in the C18-4 cells, giving the expected band of 292 bp (lane B) by RT-PCR. Fth117 expression is found in the 6-day-old testis but is not found in Sertoli cells (lanes D and F, respectively). In addition to spermatogonial-specific genes with a known function, the C18-4 cell line expressed five novel genes that belong to the Tex (testis-expressed) gene family (Fig. 6D) . None of those genes were expressed by the Sertoli cell line.

    Figure 6. Expression of spermatogonia-specific genes in the C18-4 cell line, visualized by RT-PCR. RNA samples were isolated from the C18-4 cells, from 6-day-old testes as positive control, and from the SF7 Sertoli cell line as negative control. After reverse transcription, the cDNA samples were amplified using the primers described by Wang et al. . (A): RT-PCR for Ott (PCR product = 551 and 503 bp). (B): RT-PCR for Sycp1 (PCR product = 311 bp). (C): RT-PCR for Fth117 (PCR product = 292 bp). (D): RT-PCR for Tex13 (PCR product = 220 bp), Tex14 (PCR product = 635 bp), Tex15 (PCR product = 411 bp), Tex16 (PCR product = 660 bp), and Tex19 (PCR product = 184 bp). (E): RT-PCR for Piwi1 (PCR product = 241 bp). (F): RT-PCR for Prame11 (PCR product = 448 bp). Lanes A: size markers (100 bp ladder); B: C18-4 cell line; C: negative control for C18-4; D: 6-day-old testis; E: negative control for 6-day-old testis; F: SF7 Sertoli cells; G: negative control for SF7 Sertoli cells. Abbreviation: RT-PCR, reverse transcriptase polymerase chain reaction.

    Interestingly, the C18-4 spermatogonial cell line also shows the expression of the piwi12 (piwi-like 2) gene (Fig. 6E). Piwi belongs to a family of genes that is involved in stem cell maintenance and renewal in Drosophila . Two mouse homologues of piwi have recently been cloned: miwi (mouse piwi) and mili (miwi-like) . The primers used in this study are derived from the Drosophila sequence and give a band of 241 bp. As expected, Piwi12 was also detected in the neonatal testis but not in Sertoli cells. Additionally, the C18-4 cells express a PRAME-like gene (Fig. 6F). PRAME is a protein expressed in the testis, as well as in several carcinomas, in acute myeloid leukemias, and in CD34+ hematopoietic stem cells .

    Influence of GDNF and SCF on the Behavior of the C18-4 Cells

    To determine if the C18-4 cell line responds to growth factors, we cultivated the cells in the presence or absence of 100 ng/ml GDNF in the culture medium. When GDNF is present, the rate of proliferation of the cells increases significantly (p < .005) (Fig. 7A). No influence on cell morphology or expression of the c-kit receptor was observed. Interestingly, the growth effect of GDNF is detected only when the culture medium also contains 10% FCS. No change in the rate of proliferation can be detected when FCS is replaced by a defined serum (Nu serum), which contains a minimal amount of cytokines. Further, addition of SCF to the culture medium did not stimulate the proliferation of the cells (Fig. 7B). SCF did not induce the cells to differentiate since RT-PCR analysis did not reveal the expression of LDHC4, a germ cell–specific isozyme that is detected at the earliest in preleptotene spermatocytes .

    Figure 7. Rates of proliferation of the C18-4 cells with GDNF or SCF. (A): In the presence of GDNF, the rate of proliferation of the C18-4 cells increases significantly in comparison with the control cultures. (B): No changes could be observed when the cells are cultured with SCF. Abbreviations: GDNF, glial cell line–derived neurotrophic factor; SCF, stem cell factor.

    DISCUSSION

    We thank Dr. Renee Reijo-Pera, Department of Obstetrics, Gynecology and Reproductive Sciences, University of California, San Francisco, for providing us with the Dazl no. 149 and 150 antibodies. We also thank Ms. Kathy van der Wee for valuable technical assistance and Dr. Kathy Beal for statistical analysis. This work was supported by the National Institutes of Health grant RO1-HD36483.

    REFERENCES

    Huckins C. The spermatogonial stem cell population in adult rats. I: their morphology, proliferation and maturation. Anat Rec 1971;169:533–557.

    Oakberg EF. Spermatogonial stem-cell renewal in the mouse. Anat Rec 1971;169:515–531.

    de Rooij DG, Russell LD. All you wanted to know about spermatogonia but were afraid to ask. J Androl 2000;21:776–798.

    Dym M, Fawcett DW. The blood-testis barrier in the rat and the physiological compartmentation of the seminiferous epithelium. Biol Reprod 1970;3:308–326.

    Skinner MK. Cell-cell interactions in the testis. Endocrine Rev 1991;12:45–77.

    Jegou B. The Sertoli-germ cell communication network in mammals. Int Rev Cytol 1993;147:25–96.

    Brinster RL, Zimmermann JW. Spermatogenesis following male germ-cell transplantation . Proc Natl Acad Sci U S A 1994;91:11298–11302.

    Brinster RL, Avarbock MR. Germline transmission of donor haplotype following spermatogonial transplantation . Proc Natl Acad Sci U S A 1994;91:11303–11307.

    Palombi F, Salanova M, Tarone G et al. Distribution of beta 1 integrin subunit in rat seminiferous epithelium. Biol Reprod 1992;47:1173–1182.

    Schaller J, Glander HJ, Dethloff J. Evidence of beta 1 integrins and fibronectin on spermatogenic cells in human testis. Hum Reprod 1993;8:1873–1878.

    Shinohara T, Avarbock MR, Brinster RL. ?1- and 6-integrin are surface markers on mouse spermatogonial stem cells. Proc Natl Acad Sci U S A 1999;96:5504–5509.

    Shinohara T, Orwig KE, Avarbock MR et al. Spermatogonial stem cell enrichment by multiparameter selection of mouse testis cells. Proc Natl Acad Sci U S A 2000;97:8346–8351.

    Viglietto G, Dolci S, Bruni P et al. Glial cell line-derived neutrotrophic factor and neurturin can act as paracrine growth factors stimulating DNA synthesis of Ret-expressing spermatogonia. Int J Oncol 2000;16:689–694.

    Dettin L, Ravindranath N, Hofmann MC et al. Morphological characterization of the spermatogonial subtypes in the neonatal mouse testis. Biol Reprod 2003;69:1565–1571.

    Yoshinaga K, Nishikawa S, Ogawa M et al. Role of c-kit in mouse spermatogenesis: identification of spermatogonia as a specific site of c-kit expression and function. Development 1991;113:689–699.

    Sorrentino V, Giorgi M, Geremia R et al. Expression of the c-kit proto-oncogene in the murine male germ cells. Oncogene 1991;6:149–151.

    Besmer P, Manova K, Duttlinger R et al. The kit-ligand (steel factor) and its receptor c-kit/W: pleiotropic roles in gametogenesis and melanogenesis. Development (Suppl) 1993;125–137.

    No D, Yao TP, Evans RM. Ecdysone-inducible gene expression in mammalian cells and transgenic mice. Proc Natl Acad Sci U S A 1996;93:3346–3351.

    Dym M, Jia MC, Dirami G et al. Expression of c-kit receptor and its autophosphorylation in immature rat type A spermatogonia. Biol Reprod 1995;52:8–19.

    Paquis-Flucklinger V, Michiels JF, Vidal Fetal. Expression in transgenic mice of the large T antigen of polyomavirus induces Sertoli cell tumours and allows the establishment of differentiated cell lines. Oncogene 1993;8:2087–2094.

    Hofmann MC, Narisawa S, Hess RA et al. Immortalization of germ cells and somatic testicular cells using the SV40 large T antigen. Exp Cell Res 1992;201:417–435.

    Reijo RA, Dorfman DM, Slee R et al. DAZ family proteins exist throughout male germ cell development and transit from nucleus to cytoplasm at meiosis in humans and mice. Biol Reprod 2000;63:1490–1496.

    Schrans-Stassen BH, van de Kant HJ, de Rooij DG et al. Differential expression of c-kit in mouse undifferentiated and differentiating type A spermatogonia. Endocrinology 1999;140:5894–5900.

    Min BH, Strauch AR, Foster DN. Nucleotide sequence of a mouse vascular smooth muscle -actin cDNA. Nucleic Acids Res 1988;16:10374.

    Baker PJ, Sha JA, McBride MW et al. Expression of 3?-hydroxysteroid dehydrogenase type I and type VI isoforms in the mouse testis during development. Eur J Biochem 1999;260:911–917.

    Wang PJ, McCarrey JR, Yang F et al. An abundance of X-linked genes expressed in spermatogonia. Nat Genet 2001;27:422–426.

    Gorman CM, Moffat LF, Howard BH. Recombinant genomes which express chloramphenicol acetyltransferase in mammalian cells. Mol Cell Biol 1982;2:1044–1051.

    Kerr SM, Taggart MH, Lee M et al. Ott, a mouse X-linked multigene family expressed specifically during meiosis. Hum Mol Genet 1996;5:1139–1148.

    Sage J, Martin L, Meuwissen R et al. Temporal and spatial control of the Sycp1 gene transcription in the mouse meiosis: regulatory elements active in the male are not sufficient for expression in the female gonad. Mech Dev 1999;80:29–39.

    Cox DN, Chao A, Baker J et al. A novel class of evolutionarily conserved genes defined by piwi are essential for stem cell self-renewal. Genes Dev 1998;12:3715–3727.

    Kuramochi-Miyagawa S, Kimura T, Yomogida K et al. Two mouse piwi-related genes: miwi and mili. Mech Dev 2001;108:121–133.

    Deng W, Lin H, Qiao D et al. Miwi, a murine homolog of piwi, encodes a cytoplasmic protein essential for spermatogenesis. Dev Cell 2002;2:819–830.

    Kuramochi-Miyagawa S, Kimura T, Ijiri TW et al. Mili, a mammalian member of piwi family gene, is essential for spermatogenesis. Development 2004;131:839–849.

    Kirkin AF, Dzhandzhugazyan K, Zeuthen J. Melanoma-associated antigens recognized by cytotoxic T lymphocytes. APMIS 1998;106:665–679.

    Pellat-Deceunynck C, Mellerin MP, Labarriere N et al. The cancer germ-line genes MAGE-1, MAGE-3 and PRAME are commonly expressed by human myeloma cells. Eur J Immunol 2000;30:803–809.

    Steinbach D, Hermann J, Viehmann S et al. Clinical implications of PRAME gene expression in childhood acute myeloid leukemia. Cancer Genet Cytogenet 2002;133:118–123.

    Li CM, Guo M, Borczuk A et al. Gene expression in Wilms’ tumor mimics the earliest committed stage in the metanephric mesenchymal-epithelial transition. Am J Pathol 2002;160:2181–2190.

    Li SS, O’Brien DA, Hou EW et al. Differential activity and synthesis of lactate dehydrogenase isozymes A (muscle), B (heart), and C (testis) in mouse spermatogenic cells. Biol Reprod 1989;40:173–180.

    Alcivar AA, Trasler JM, Hake LE et al. DNA methylation and expression of the genes coding for lactate dehydrogenases A and C during rodent spermatogenesis. Biol Reprod 1991;44:527–535.

    Cooke HJ, Lee M, Kerr S et al. A murine homologue of the human DAZ gene is autosomal and expressed only in male and female gonads. Hum Mol Genet 1996;5:513–516.

    Reijo R, Seligman J, Dinulos MB et al. Mouse autosomal homolog of DAZ, a candidate male sterility gene in humans, is expressed in male germ cells before and after puberty. Genomics 1996;35:346–352.

    Ruggiu M, Speed R, Taggart M et al. The mouse Dazla gene encodes a cytoplasmic protein essential for gametogenesis. Nature 1997;389:73–77.

    Jing S, Wen D, Yu Y et al. GDNF-induced activation of the ret protein tyrosine kinase is mediated by GDNFR-alpha, a novel receptor for GDNF. Cell 1996;85:1113–1124.

    Creemers LB, Meng X, den OK et al. Transplantation of germ cells from glial cell line-derived neurotrophic factor-overexpressing mice to host testes depleted of endogenous spermatogenesis by fractionated irradiation. Biol Reprod 2002;66:1579–1584.

    Ruggiu M, Cooke HJ. In vivo and in vitro analysis of homodimerisation activity of the mouse Daz11 protein. Gene 2000;252:119–126.

    Parfenov VN, Pochukalina GN, Davis DS et al. Nuclear distribution of Oct-4 transcription factor in transcriptionally active and inactive mouse oocytes and its relation to RNA polymerase II and splicing factors. J Cell Biochem 2003;89:720–732.

    Meng X, Lindahl M, Hyvonen ME et al. Regulation of cell fate decision of undifferentiated spermatogonia by GDNF. Science 2000;287:1489–1493.

    Meng X, de RDG, Westerdahl K et al. Promotion of seminomatous tumors by targeted overexpression of glial cell line-derived neurotrophic factor in mouse testis. Cancer Res 2001;61:3267–3271.

    Trupp M, Ryden M, Jornvall H et al. Peripheral expression and biological activities of GDNF, a new neurotrophic factor for avian and mammalian peripheral neurons. J Cell Biol 1995;130:137–148.

    Golden JP, DeMaro JA, Osborne PA et al. Expression of neurturin, GDNF, and GDNF family-receptor mRNA in the developing and mature mouse. Exp Neurol 1999;158:504–528.

    Ohta H, Yomogida K, Dohmae K et al. Regulation of proliferation and differentiation in spermatogonial stem cells: the role of c-kit and its ligand SCF. Development 2000;127:2125–2131.

    Cox DN, Chao A, Lin H. Piwi encodes a nucleoplasmic factor whose activity modulates the number and division rate of germline stem cells. Development 2000;127:503–514.

    Tabara H, Sarkissian M, Kelly WG et al. The rde-1 gene, RNA interference, and transposon silencing in C. elegans. Cell 1999;99:123–132.

    Qiao D, Zeeman AM, Deng W et al. Molecular characterization of hiwi, a human member of the piwi gene family whose overexpression is correlated to seminomas. Oncogene 2002;21:3988–3999.

    Scanlan MJ, Gure AO, Jungbluth AA et al. Cancer/testis antigens: an expanding family of targets for cancer immunotherapy. Immunol Rev 2002;188:22–32.

    Feng LX, Chen Y, Dettin L et al. Generation and in vitro differentiation of a spermatogonial cell line. Science 2002;297:392–395.

    van Pelt AM, Roepers-Gajadien HL, Gademan IS et al. Establishment of cell lines with rat spermatogonial stem cell characteristics. Endocrinology 2002;143:1845–1850.

    Krieglstein K, Henheik P, Farkas L et al. Glial cell line-derived neurotrophic factor requires transforming growth factor-beta for exerting its full neurotrophic potential on peripheral and CNS neurons. J Neurosci 1998;18:9822–9834.

    Kanatsu-Shinohara M, Ogonuki N, Inoue K et al. Long-term proliferation in culture and germline transmission of mouse male germline stem cells. Biol Reprod 2003;69:612–616.(Marie-Claude Hofmanna, La)