Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

Genetics of germ cell development

Key Points

  • Germ cells are specialized cells that are responsible for transmitting the genome of an individual organism to its offspring.

  • The defining characteristic of the germ cells is their ability to undergo meiosis, in which the diploid genome is reduced to a haploid genome that can combine with another haploid genome at fertilization.

  • Many of the factors specifying germ cell identity are RNA-binding factors, and many of these RNA-binding factors are conserved in the germ cells across multiple species.

  • Maintenance of a transcriptionally repressed state is a characteristic of early germ cells in multiple species. Repression is accomplished both by the direct inhibition of RNA polymerase II and by the establishment of a repressive chromatin configuration.

  • The decision to stop mitotic proliferation and to enter meiosis is timed differently in the different species and in different sexes of the same species. In some cases, a proliferative pool of germline precursors is retained after this decision, and in some cases all available germ cells enter meiosis together.

  • The later steps of germ cell development set up the cues that will guide the earliest stages of embryogenesis.

  • Germ cells represent the closest in vivo equivalent to in vitro pluripotent stem cell systems; understanding germ cell biology will provide new insights into the nature of pluripotency.

Abstract

The germ line represents a continuous cellular link between generations and between species, but the germ cells themselves develop in a specialized, organism-specific context. The model organisms Caenorhabditis elegans, Drosophila melanogaster and the mouse display striking similarities, as well as major differences, in the means by which they control germ cell development. Recent developments in genetic technologies allow a more detailed comparison of the germ cells of these three organisms than has previously been possible, shedding light not only on universal aspects of germline regulation, but also on the control of the pluripotent state in vivo and on the earliest steps of embryogenesis. Here, we highlight themes from the comparison of these three alternative strategies for navigating the fundamental cycle of sexual reproduction.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Summary of germ cell development in C. elegans, D. melanogaster and the mouse
Figure 2: Mechanism of transcriptional repression by PIE-1 and PGC.
Figure 3: Genetic pathways controlling germ cell sex-determination and meiotic entry.
Figure 4: Meiotic arrest in developing oocytes.

References

  1. Richardson, B. E. & Lehmann, R. Mechanisms guiding primordial germ cell migration: strategies from different organisms. Nature Rev. Mol. Cell Biol. 11, 37–49 (2010).

    Article  CAS  Google Scholar 

  2. Von Stetina, J. R. & Orr-Weaver, T. L. Developmental control of oocyte maturation and egg activation in metazoan models. Cold Spring Harb. Perspect. Biol. 3, a005553 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Yanowitz, J. Meiosis: making a break for it. Curr. Opin. Cell Biol. 22, 744–751 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Hermo, L., Pelletier, R. M., Cyr, D. G. & Smith, C. E. Surfing the wave, cycle, life history, and genes/proteins expressed by testicular germ cells. Part 2: changes in spermatid organelles associated with development of spermatozoa. Microsc. Res. Tech. 73, 279–319 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Extavour, C. G. & Akam, M. Mechanisms of germ cell specification across the metazoans: epigenesis and preformation. Development 130, 5869–5884 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Updike, D. & Strome, S. P granule assembly and function in Caenorhabditis elegans germ cells. J. Androl. 31, 53–60 (2010).

    Article  CAS  PubMed  Google Scholar 

  7. Strome, S. & Wood, W. B. Immunofluorescence visualization of germ-line-specific cytoplasmic granules in embryos, larvae, and adults of Caenorhabditis elegans. Proc. Natl Acad. Sci. USA 79, 1558–1562 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gallo, C. M., Wang, J. T., Motegi, F. & Seydoux, G. Cytoplasmic partitioning of P granule components is not required to specify the germline in C. elegans. Science 330, 1685–1689 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Illmensee, K. & Mahowald, A. P. Transplantation of posterior polar plasm in Drosophila. Induction of germ cells at the anterior pole of the egg. Proc. Natl Acad. Sci. USA 71, 1016–1020 (1974).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lehmann, R. & Nusslein-Volhard, C. Abdominal segmentation, pole cell formation, and embryonic polarity require the localized activity of oskar, a maternal gene in Drosophila. Cell 47, 141–152 (1986).

    Article  CAS  PubMed  Google Scholar 

  11. Ephrussi, A. & Lehmann, R. Induction of germ cell formation by oskar. Nature 358, 387–392 (1992). The authors show that a specific gene, oskar , is sufficient to induce the formation of germ plasm and the development of functional germ cells in an ectopic location.

    Article  CAS  PubMed  Google Scholar 

  12. Thomson, T. & Lasko, P. Drosophila tudor is essential for polar granule assembly and pole cell specification, but not for posterior patterning. Genesis 40, 164–170 (2004).

    Article  CAS  PubMed  Google Scholar 

  13. Wang, C., Dickinson, L. K. & Lehmann, R. Genetics of nanos localization in Drosophila. Dev. Dyn. 199, 103–115 (1994).

    Article  CAS  PubMed  Google Scholar 

  14. Lehmann, R. & Nusslein-Volhard, C. The maternal gene nanos has a central role in posterior pattern formation of the Drosophila embryo. Development 112, 679–691 (1991).

    CAS  PubMed  Google Scholar 

  15. Barbee, S. A. & Evans, T. C. The Sm proteins regulate germ cell specification during early C. elegans embryogenesis. Dev. Biol. 291, 132–143 (2006).

    Article  CAS  PubMed  Google Scholar 

  16. Gonsalvez, G. B., Rajendra, T. K., Wen, Y., Praveen, K. & Matera, A. G. Sm proteins specify germ cell fate by facilitating oskar mRNA localization. Development 137, 2341–2351 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gonsalvez, G. B., Rajendra, T. K., Tian, L. & Matera, A. G. The Sm-protein methyltransferase, dart5, is essential for germ-cell specification and maintenance. Curr. Biol. 16, 1077–1089 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Ohinata, Y. et al. Blimp1 is a critical determinant of the germ cell lineage in mice. Nature 436, 207–213 (2005).

    Article  CAS  PubMed  Google Scholar 

  19. Yamaji, M. et al. Critical function of Prdm14 for the establishment of the germ cell lineage in mice. Nature Genet. 40, 1016–1022 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Saitou, M., Barton, S. C. & Surani, M. A. A molecular programme for the specification of germ cell fate in mice. Nature 418, 293–300 (2002).

    Article  CAS  PubMed  Google Scholar 

  21. Tanaka, S. S. & Matsui, Y. Developmentally regulated expression of mil-1 and mil-2, mouse interferon-induced transmembrane protein like genes, during formation and differentiation of primordial germ cells. Mech. Dev. 119, S261–S267 (2002).

    Article  PubMed  Google Scholar 

  22. Tam, P. P. & Zhou, S. X. The allocation of epiblast cells to ectodermal and germ-line lineages is influenced by the position of the cells in the gastrulating mouse embryo. Dev. Biol. 178, 124–132 (1996). The authors transplanted epiblast cells to heterotopic sites in developing mouse embryos, demonstrating that position, rather than cell lineage, determines the presumptive germ line in mouse.

    Article  CAS  PubMed  Google Scholar 

  23. Ohinata, Y. et al. A signaling principle for the specification of the germ cell lineage in mice. Cell 137, 571–584 (2009).

    Article  CAS  PubMed  Google Scholar 

  24. de Sousa Lopes, S. M. et al. BMP signaling mediated by ALK2 in the visceral endoderm is necessary for the generation of primordial germ cells in the mouse embryo. Genes Dev. 18, 1838–1849 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Ying, Y., Liu, X. M., Marble, A., Lawson, K. A. & Zhao, G. Q. Requirement of Bmp8b for the generation of primordial germ cells in the mouse. Mol. Endocrinol. 14, 1053–1063 (2000).

    Article  CAS  PubMed  Google Scholar 

  26. Ying, Y. & Zhao, G. Q. Cooperation of endoderm-derived BMP2 and extraembryonic ectoderm-derived BMP4 in primordial germ cell generation in the mouse. Dev. Biol. 232, 484–492 (2001).

    Article  CAS  PubMed  Google Scholar 

  27. Yamaguchi, S., Kimura, H., Tada, M., Nakatsuji, N. & Tada, T. Nanog expression in mouse germ cell development. Gene Expr. Patterns 5, 639–646 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Yabuta, Y., Kurimoto, K., Ohinata, Y., Seki, Y. & Saitou, M. Gene expression dynamics during germline specification in mice identified by quantitative single-cell gene expression profiling. Biol. Reprod. 75, 705–716 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Rosner, M. H. et al. A POU-domain transcription factor in early stem cells and germ cells of the mammalian embryo. Nature 345, 686–692 (1990).

    Article  CAS  PubMed  Google Scholar 

  30. Bortvin, A., Goodheart, M., Liao, M. & Page, D. C. Dppa3 / Pgc7 / stella is a maternal factor and is not required for germ cell specification in mice. BMC Dev. Biol. 4, 2 (2004).

    Article  PubMed  PubMed Central  Google Scholar 

  31. West, J. A. et al. A role for Lin28 in primordial germ-cell development and germ-cell malignancy. Nature 460, 909–913 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Okamura, D., Tokitake, Y., Niwa, H. & Matsui, Y. Requirement of Oct3/4 function for germ cell specification. Dev. Biol. 317, 576–584 (2008).

    Article  CAS  PubMed  Google Scholar 

  33. Molyneaux, K. A., Stallock, J., Schaible, K. & Wylie, C. Time-lapse analysis of living mouse germ cell migration. Dev. Biol. 240, 488–498 (2001). Using striking movies of fluorescently labelled live germ cells in the mouse embryo, this paper demonstrated that germ cells actively migrate from the gut into the genital ridge.

    Article  CAS  PubMed  Google Scholar 

  34. Jaglarz, M. K. & Howard, K. R. Primordial germ cell migration in Drosophila melanogaster is controlled by somatic tissue. Development 120, 83–89 (1994).

    CAS  PubMed  Google Scholar 

  35. Seydoux, G. et al. Repression of gene expression in the embryonic germ lineage of C. elegans. Nature 382, 713–716 (1996).

    Article  CAS  PubMed  Google Scholar 

  36. Mello, C. C. et al. The PIE-1 protein and germline specification in C. elegans embryos. Nature 382, 710–712 (1996). References 35 and 36 showed that the PIE-1 protein is responsible for maintaining transcriptional repression in cells of the germline lineage in C. elegans.

    Article  CAS  PubMed  Google Scholar 

  37. Batchelder, C. et al. Transcriptional repression by the Caenorhabditis elegans germ-line protein PIE-1. Genes Dev. 13, 202–212 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Zhang, F., Barboric, M., Blackwell, T. K. & Peterlin, B. M. A model of repression: CTD analogs and PIE-1 inhibit transcriptional elongation by P-TEFb. Genes Dev. 17, 748–758 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Ghosh, D. & Seydoux, G. Inhibition of transcription by the Caenorhabditis elegans germline protein PIE-1: genetic evidence for distinct mechanisms targeting initiation and elongation. Genetics 178, 235–243 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Nakamura, A., Amikura, R., Mukai, M., Kobayashi, S. & Lasko, P. F. Requirement for a noncoding RNA in Drosophila polar granules for germ cell establishment. Science 274, 2075–2079 (1996).

    Article  CAS  PubMed  Google Scholar 

  41. Deshpande, G., Calhoun, G. & Schedl, P. Overlapping mechanisms function to establish transcriptional quiescence in the embryonic Drosophila germline. Development 131, 1247–1257 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Martinho, R. G., Kunwar, P. S., Casanova, J. & Lehmann, R. A noncoding RNA is required for the repression of RNApolII-dependent transcription in primordial germ cells. Curr. Biol. 14, 159–165 (2004).

    Article  CAS  PubMed  Google Scholar 

  43. Hanyu-Nakamura, K., Sonobe-Nojima, H., Tanigawa, A., Lasko, P. & Nakamura, A. Drosophila Pgc protein inhibits P-TEFb recruitment to chromatin in primordial germ cells. Nature 451, 730–733 (2008). This paper showed for the first time that the pgc gene encodes a protein that functions by preventing PTEFb recruitment. Prior to this publication, pgc was known to act by preventing transcription initiation but was thought to encode a non-coding RNA.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Seki, Y. et al. Cellular dynamics associated with the genome-wide epigenetic reprogramming in migrating primordial germ cells in mice. Development 134, 2627–2638 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Schaner, C. E., Deshpande, G., Schedl, P. D. & Kelly, W. G. A conserved chromatin architecture marks and maintains the restricted germ cell lineage in worms and flies. Dev. Cell. 5, 747–757 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Bender, L. B., Cao, R., Zhang, Y. & Strome, S. The MES-2/MES-3/MES-6 complex and regulation of histone H3 methylation in C. elegans. Curr. Biol. 14, 1639–1643 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Korf, I., Fan, Y. & Strome, S. The Polycomb group in Caenorhabditis elegans and maternal control of germline development. Development 125, 2469–2478 (1998).

    CAS  PubMed  Google Scholar 

  48. Holdeman, R., Nehrt, S. & Strome, S. MES-2, a maternal protein essential for viability of the germline in Caenorhabditis elegans, is homologous to a Drosophila Polycomb group protein. Development 125, 2457–2467 (1998).

    CAS  PubMed  Google Scholar 

  49. Katz, D. J., Edwards, T. M., Reinke, V. & Kelly, W. G. A. C. elegans LSD1 demethylase contributes to germline immortality by reprogramming epigenetic memory. Cell 137, 308–320 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Rudolph, T. et al. Heterochromatin formation in Drosophila is initiated through active removal of H3K4 methylation by the LSD1 homolog SU(VAR)3-3. Mol. Cell. 26, 103–115 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Kurimoto, K. et al. Complex genome-wide transcription dynamics orchestrated by Blimp1 for the specification of the germ cell lineage in mice. Genes Dev. 22, 1617–1635 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Ancelin, K. et al. Blimp1 associates with Prmt5 and directs histone arginine methylation in mouse germ cells. Nature Cell Biol. 8, 623–630 (2006).

    Article  CAS  PubMed  Google Scholar 

  53. Hirsh, D., Oppenheim, D. & Klass, M. Development of the reproductive system of Caenorhabditis elegans. Dev. Biol. 49, 200–219 (1976).

    Article  CAS  PubMed  Google Scholar 

  54. Austin, J. & Kimble, J. glp-1 is required in the germ line for regulation of the decision between mitosis and meiosis in C. elegans. Cell 51, 589–599 (1987). This study identified a molecular signal, GLP-1, that is responsible for transducing the mitosis-promoting signal from the distal tip cell to the germ cells in C. elegans , thus providing a molecular correlate for the interaction between germ cells and their niche.

    Article  CAS  PubMed  Google Scholar 

  55. Henderson, S. T. Gao, D., Lambie, E. J. & Kimble, J. lag-2 may encode a signaling ligand for the GLP-1 and LIN-12 receptors of C. elegans. Development 120, 2913–2924 (1994).

    CAS  PubMed  Google Scholar 

  56. Berry, L. W., Westlund, B. & Schedl, T. Germ-line tumor formation caused by activation of glp-1, a Caenorhabditis elegans member of the Notch family of receptors. Development 124, 925–936 (1997).

    CAS  PubMed  Google Scholar 

  57. Crittenden, S. L. et al. A conserved RNA-binding protein controls germline stem cells in Caenorhabditis elegans. Nature 417, 660–663 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Eckmann, C. R., Crittenden, S. L., Suh, N. & Kimble, J. GLD-3 and control of the mitosis/meiosis decision in the germline of Caenorhabditis elegans. Genetics 168, 147–160 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Hansen, D., Wilson-Berry, L., Dang, T. & Schedl, T. Control of the proliferation versus meiotic development decision in the C. elegans germline through regulation of GLD-1 protein accumulation. Development 131, 93–104 (2004).

    Article  CAS  PubMed  Google Scholar 

  60. Eckmann, C. R., Kraemer, B., Wickens, M. & Kimble, J. GLD-3, a bicaudal-C homolog that inhibits FBF to control germline sex determination in C. elegans. Dev. Cell. 3, 697–710 (2002).

    Article  CAS  PubMed  Google Scholar 

  61. Kraemer, B. et al. NANOS-3 and FBF proteins physically interact to control the sperm-oocyte switch in Caenorhabditis elegans. Curr. Biol. 9, 1009–1018 (1999).

    Article  CAS  PubMed  Google Scholar 

  62. Ellis, R. E. Sex determination in the Caenorhabditis elegans germ line. Curr. Top. Dev. Biol. 83, 41–64 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Kimble, J. & Crittenden, S. L. Controls of germline stem cells, entry into meiosis, and the sperm/oocyte decision in Caenorhabditis elegans. Annu. Rev. Cell Dev. Biol. 23, 405–433 (2007).

    Article  CAS  PubMed  Google Scholar 

  64. Chen, P. J., Singal, A., Kimble, J. & Ellis, R. E. A novel member of the tob family of proteins controls sexual fate in Caenorhabditis elegans germ cells. Dev. Biol. 217, 77–90 (2000).

    Article  CAS  PubMed  Google Scholar 

  65. Chen, P. & Ellis, R. E. TRA-1A regulates transcription of fog-3, which controls germ cell fate in C. elegans. Development 127, 3119–3129 (2000).

    CAS  PubMed  Google Scholar 

  66. Barton, M. K. & Kimble, J. fog-1, a regulatory gene required for specification of spermatogenesis in the germ line of Caenorhabditis elegans. Genetics 125, 29–39 (1990).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Ellis, R. E. & Kimble, J. The fog-3 gene and regulation of cell fate in the germ line of Caenorhabditis elegans. Genetics 139, 561–577 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Thompson, B. E. et al. Dose-dependent control of proliferation and sperm specification by FOG-1/CPEB. Development 132, 3471–3481 (2005).

    Article  CAS  PubMed  Google Scholar 

  69. Hodgkin, J. Sex determination in the nematode C. elegans: analysis of tra-3 suppressors and characterization of fem genes. Genetics 114, 15–52 (1986).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Starostina, N. G. et al. A CUL-2 ubiquitin ligase containing three FEM proteins degrades TRA-1 to regulate C. elegans sex determination. Dev. Cell. 13, 127–139 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Otori, M., Karashima, T. & Yamamoto, M. The Caenorhabditis elegans homologue of deleted in azoospermia is involved in the sperm/oocyte switch. Mol. Biol. Cell. 17, 3147–3155 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Zhang, B. et al. A conserved RNA-binding protein that regulates sexual fates in the C. elegans hermaphrodite germ line. Nature 390, 477–484 (1997).

    Article  CAS  PubMed  Google Scholar 

  73. Williamson, A. & Lehmann, R. Germ cell development in Drosophila. Annu. Rev. Cell Dev. Biol. 12, 365–391 (1996).

    Article  CAS  PubMed  Google Scholar 

  74. Su, T. T., Campbell, S. D. amp; O'Farrell, P. H. The cell cycle program in germ cells of the Drosophila embryo. Dev. Biol. 196, 160–170 (1998).

    Article  CAS  PubMed  Google Scholar 

  75. Zalokar, M. Autoradiographic study of protein and RNA formation during early development of Drosophila eggs. Dev. Biol. 49, 425–437 (1976).

    Article  CAS  PubMed  Google Scholar 

  76. Hashiyama, K., Hayashi, Y. & Kobayashi, S. Drosophila Sex lethal gene initiates female development in germline progenitors. Science 333, 885–888 (2011).

    Article  CAS  PubMed  Google Scholar 

  77. Casper, A. L. & Van Doren, M. The establishment of sexual identity in the Drosophila germline. Development 136, 3821–3830 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pauli, D., Oliver, B. & Mahowald, A. P. The role of the ovarian tumor locus in Drosophila melanogaster germ line sex determination. Development 119, 123–134 (1993).

    CAS  PubMed  Google Scholar 

  79. Lu, J. & Oliver, B. Drosophila OVO regulates ovarian tumor transcription by binding unusually near the transcription start site. Development 128, 1671–1686 (2001).

    CAS  PubMed  Google Scholar 

  80. Wawersik, M. et al. Somatic control of germline sexual development is mediated by the JAK/STAT pathway. Nature 436, 563–567 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. King, F. J. & Lin, H. Somatic signaling mediated by fs(1)Yb is essential for germline stem cell maintenance during Drosophila oogenesis. Development 126, 1833–1844 (1999).

    CAS  PubMed  Google Scholar 

  82. Demerec, M. Biology of Drosophila, (John Wiley & Sons, 1950).

    Google Scholar 

  83. Wang, Z. & Lin, H. Nanos maintains germline stem cell self-renewal by preventing differentiation. Science 303, 2016–2019 (2004).

    Article  CAS  PubMed  Google Scholar 

  84. Gilboa, L. & Lehmann, R. Repression of primordial germ cell differentiation parallels germ line stem cell maintenance. Curr. Biol. 14, 981–986 (2004).

    Article  CAS  PubMed  Google Scholar 

  85. Xie, T. & Spradling, A. C. decapentaplegic is essential for the maintenance and division of germline stem cells in the Drosophila ovary. Cell 94, 251–260 (1998).

    Article  CAS  PubMed  Google Scholar 

  86. Song, X. et al. Bmp signals from niche cells directly repress transcription of a differentiation-promoting gene, bag of marbles, in germline stem cells in the Drosophila ovary. Development 131, 1353–1364 (2004).

    Article  CAS  PubMed  Google Scholar 

  87. Kiger, A. A., Jones, D. L., Schulz, C., Rogers, M. B. & Fuller, M. T. Stem cell self-renewal specified by JAK-STAT activation in response to a support cell cue. Science 294, 2542–2545 (2001).

    Article  CAS  PubMed  Google Scholar 

  88. Lavoie, C. A., Ohlstein, B. & McKearin, D. M. Localization and function of Bam protein require the benign gonial cell neoplasm gene product. Dev. Biol. 212, 405–413 (1999).

    Article  CAS  PubMed  Google Scholar 

  89. Gonczy, P., Matunis, E. & DiNardo, S. bag-of-marbles and benign gonial cell neoplasm act in the germline to restrict proliferation during Drosophila spermatogenesis. Development 124, 4361–4371 (1997).

    CAS  PubMed  Google Scholar 

  90. Li, Y., Minor, N. T., Park, J. K., McKearin, D. M. & Maines, J. Z. Bam and Bgcn antagonize Nanos-dependent germ-line stem cell maintenance. Proc. Natl Acad. Sci. USA 106, 9304–9309 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Bastock, R. & St Johnston, D. Drosophila oogenesis. Curr. Biol. 18, R1082–R1087 (2008).

    Article  CAS  PubMed  Google Scholar 

  92. Fuller, M. T. Genetic control of cell proliferation and differentiation in Drosophila spermatogenesis. Semin. Cell Dev. Biol. 9, 433–444 (1998).

    Article  CAS  PubMed  Google Scholar 

  93. Huynh, J. R. & St Johnston, D. The role of BicD, Egl, Orb and the microtubules in the restriction of meiosis to the Drosophila oocyte. Development 127, 2785–2794 (2000).

    CAS  PubMed  Google Scholar 

  94. Tam, P. P. & Snow, M. H. Proliferation and migration of primordial germ cells during compensatory growth in mouse embryos. J. Embryol. Exp. Morphol. 64, 133–147 (1981).

    CAS  PubMed  Google Scholar 

  95. Hajkova, P. et al. Epigenetic reprogramming in mouse primordial germ cells. Mech. Dev. 117, 15–23 (2002).

    Article  CAS  PubMed  Google Scholar 

  96. Tanaka, S. S. et al. The mouse homolog of Drosophila Vasa is required for the development of male germ cells. Genes Dev. 14, 841–853 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Seligman, J. & Page, D. C. The Dazh gene is expressed in male and female embryonic gonads before germ cell sex differentiation. Biochem. Biophys. Res. Commun. 245, 878–882 (1998).

    Article  CAS  PubMed  Google Scholar 

  98. Enders, G. C. & May, J. J. Developmentally regulated expression of a mouse germ cell nuclear antigen examined from embryonic day 11 to adult in male and female mice. Dev. Biol. 163, 331–340 (1994).

    Article  CAS  PubMed  Google Scholar 

  99. Gill, M. E., Hu, Y. C., Lin, Y. & Page, D. C. Licensing of gametogenesis, dependent on RNA binding protein DAZL, as a gateway to sexual differentiation of fetal germ cells. Proc. Natl Acad. Sci. USA 108, 7443–7448 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Lin, Y., Gill, M. E., Koubova, J. & Page, D. C. Germ cell-intrinsic and -extrinsic factors govern meiotic initiation in mouse embryos. Science 322, 1685–1687 (2008). This is the first paper demonstrating the existence of a 'licensing' step during mammalian germ cell development, and shows that the Dazl gene is required for licensing.

    Article  CAS  PubMed  Google Scholar 

  101. Hilscher, B. et al. Kinetics of gametogenesis. I. Comparative histological and autoradiographic studies of oocytes and transitional prospermatogonia during oogenesis and prespermatogenesis. Cell Tissue Res. 154, 443–470 (1974).

    Article  CAS  PubMed  Google Scholar 

  102. Baltus, A. E. et al. In germ cells of mouse embryonic ovaries, the decision to enter meiosis precedes premeiotic DNA replication. Nature Genet. 38, 1430–1434 (2006).

    Article  CAS  PubMed  Google Scholar 

  103. Anderson, E. L. et al. Stra8 and its inducer, retinoic acid, regulate meiotic initiation in both spermatogenesis and oogenesis in mice. Proc. Natl Acad. Sci. USA 105, 14976–14980 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Menke, D. B., Koubova, J. & Page, D. C. Sexual differentiation of germ cells in XX mouse gonads occurs in an anterior-to-posterior wave. Dev. Biol. 262, 303–312 (2003).

    Article  CAS  PubMed  Google Scholar 

  105. Koubova, J. et al. Retinoic acid regulates sex-specific timing of meiotic initiation in mice. Proc. Natl Acad. Sci. USA 103, 2474–2479 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Bowles, J. et al. Retinoid signaling determines germ cell fate in mice. Science 312, 596–600 (2006). References 102, 105 and 106 demonstrate that retinoic acid– Stra8 signalling is necessary and sufficient for the initiation of meiosis in mammalian germ cells. They also show that degradation of this signal in male embryos is responsible for the sex-specific timing of meiotic initiation.

    Article  CAS  PubMed  Google Scholar 

  107. Tsuda, M. et al. Conserved role of nanos proteins in germ cell development. Science 301, 1239–1241 (2003).

    Article  CAS  PubMed  Google Scholar 

  108. Suzuki, A. & Saga, Y. Nanos2 suppresses meiosis and promotes male germ cell differentiation. Genes Dev. 22, 430–435 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Jameson, S. A. et al. Temporal transcriptional profiling of somatic and germ cells reveals biased lineage priming of sexual fate in the fetal mouse gonad. PLoS Genet. 8, e1002575 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. De Leon, V., Johnson, A. & Bachvarova, R. Half-lives and relative amounts of stored and polysomal ribosomes and poly(A)+ RNA in mouse oocytes. Dev. Biol. 98, 400–408 (1983).

    Article  CAS  PubMed  Google Scholar 

  111. L'Hernault, S. W. Spermatogenesis. in WormBook: The Online Review of C. elegans Biology (2006).

  112. Wolf, N., Hirsh, D. & McIntosh, J. R. Spermatogenesis in males of the free-living nematode, Caenorhabditis elegans. J. Ultrastruct. Res. 63, 155–169 (1978).

    Article  CAS  PubMed  Google Scholar 

  113. Ooi, S. L., Priess, J. R. & Henikoff, S. Histone H3.3 variant dynamics in the germline of Caenorhabditis elegans. PLoS Genet. 2, e97 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Miller, M. A. et al. A sperm cytoskeletal protein that signals oocyte meiotic maturation and ovulation. Science 291, 2144–2147 (2001). The authors describe the unexpected finding that a single protein has both a structural function in the sperm mobility apparatus, and a hormonal function in inducing ovulation and releasing meiotic arrest in the oocytes.

    Article  CAS  PubMed  Google Scholar 

  115. Arico, J. K., Katz, D. J., van der Vlag, J. & Kelly, W. G. Epigenetic patterns maintained in early Caenorhabditis elegans embryos can be established by gene activity in the parental germ cells. PLoS Genet. 7, e1001391 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Karashima, T., Sugimoto, A. & Yamamoto, M. Caenorhabditis elegans homologue of the human azoospermia factor DAZ is required for oogenesis but not for spermatogenesis. Development 127, 1069–1079 (2000).

    CAS  PubMed  Google Scholar 

  117. Francis, R., Maine, E. & Schedl, T. Analysis of the multiple roles of gld-1 in germline development: interactions with the sex determination cascade and the glp-1 signaling pathway. Genetics 139, 607–630 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. McNally, K. L. & McNally, F. J. Fertilization initiates the transition from anaphase I to metaphase II during female meiosis in C. elegans. Dev. Biol. 282, 218–230 (2005).

    Article  CAS  PubMed  Google Scholar 

  119. Goldstein, B. & Hird, S. N. Specification of the anteroposterior axis in Caenorhabditis elegans. Development 122, 1467–1474 (1996).

    CAS  PubMed  Google Scholar 

  120. Eberhart, C. G., Maines, J. Z. & Wasserman, S. A. Meiotic cell cycle requirement for a fly homologue of human Deleted in Azoospermia. Nature 381, 783–785 (1996).

    Article  CAS  PubMed  Google Scholar 

  121. Gonzalez-Reyes, A., Elliott, H. & St Johnston, D. Polarization of both major body axes in Drosophila by gurken-torpedo signalling. Nature 375, 654–658 (1995).

    Article  CAS  PubMed  Google Scholar 

  122. Bonnefoy, E., Orsi, G. A., Couble, P. & Loppin, B. The essential role of Drosophila HIRA for de novo assembly of paternal chromatin at fertilization. PLoS Genet. 3, 1991–2006 (2007).

    Article  CAS  PubMed  Google Scholar 

  123. Adenot, P. G., Mercier, Y., Renard, J. P. & Thompson, E. M. Differential H4 acetylation of paternal and maternal chromatin precedes DNA replication and differential transcriptional activity in pronuclei of 1-cell mouse embryos. Development 124, 4615–4625 (1997).

    CAS  PubMed  Google Scholar 

  124. Tsukamoto, S. et al. Autophagy is essential for preimplantation development of mouse embryos. Science 321, 117–120 (2008).

    Article  CAS  PubMed  Google Scholar 

  125. Roest, H. P. et al. The ubiquitin-conjugating DNA repair enzyme HR6A is a maternal factor essential for early embryonic development in mice. Mol. Cell. Biol. 24, 5485–5495 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Lasko, P. Posttranscriptional regulation in Drosophila oocytes and early embryos. Wiley Interdiscip. Rev. RNA 2, 408–416 (2011).

    Article  CAS  PubMed  Google Scholar 

  127. Kershner, A. M. & Kimble, J. Genome-wide analysis of mRNA targets for Caenorhabditis elegans FBF, a conserved stem cell regulator. Proc. Natl Acad. Sci. USA 107, 3936–3941 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Qiu, C. et al. Divergence of Pumilio/fem-3 mRNA binding factor (PUF) protein specificity through variations in an RNA-binding pocket. J. Biol. Chem. 287, 6949–6957 (2012).

    Article  CAS  PubMed  Google Scholar 

  129. Saxe, J. P. & Lin, H. Small noncoding RNAs in the germline. Cold Spring Harb. Perspect. Biol. 3, a002717 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Banaszynski, L. A., Allis, C. D. & Lewis, P. W. Histone variants in metazoan development. Dev. Cell. 19, 662–674 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Hardy, R. W., Tokuyasu, K. T., Lindsley, D. L. & Garavito, M. The germinal proliferation center in the testis of Drosophila melanogaster. J. Ultrastruct. Res. 69, 180–190 (1979).

    Article  CAS  PubMed  Google Scholar 

  132. Kimble, J. E. & White, J. G. On the control of germ cell development in Caenorhabditis elegans. Dev. Biol. 81, 208–219 (1981).

    Article  CAS  PubMed  Google Scholar 

  133. Yoshida, S., Sukeno, M. & Nabeshima, Y. A vasculature-associated niche for undifferentiated spermatogonia in the mouse testis. Science 317, 1722–1726 (2007).

    Article  CAS  PubMed  Google Scholar 

  134. Labosky, P. A., Barlow, D. P. & Hogan, B. L. Mouse embryonic germ (EG) cell lines: transmission through the germline and differences in the methylation imprint of insulin-like growth factor 2 receptor (Igf2r) gene compared with embryonic stem (ES) cell lines. Development 120, 3197–3204 (1994).

    CAS  PubMed  Google Scholar 

  135. Damjanov, I. The road from teratocarcinoma to human embryonic stem cells. Stem Cell Rev. 1, 273–276 (2005).

    Article  PubMed  Google Scholar 

  136. Nayernia, K. et al. In vitro-differentiated embryonic stem cells give rise to male gametes that can generate offspring mice. Dev. Cell. 11, 125–132 (2006).

    Article  CAS  PubMed  Google Scholar 

  137. Hubner, K. et al. Derivation of oocytes from mouse embryonic stem cells. Science 300, 1251–1256 (2003).

    Article  CAS  PubMed  Google Scholar 

  138. Hayashi, K., Ohta, H., Kurimoto, K., Aramaki, S. & Saitou, M. Reconstitution of the mouse germ cell specification pathway in culture by pluripotent stem cells. Cell. 146, 519–532 (2011). This study is the first to describe the production of healthy, fertile offspring from mouse spermatogenic precursors produced in vitro.

    Article  CAS  PubMed  Google Scholar 

  139. Geijsen, N. et al. Derivation of embryonic germ cells and male gametes from embryonic stem cells. Nature 427, 148–154 (2004).

    Article  CAS  PubMed  Google Scholar 

  140. Greer, E. L. et al. Transgenerational epigenetic inheritance of longevity in Caenorhabditis elegans. Nature 479, 365–371 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Seong, K. H., Li, D., Shimizu, H., Nakamura, R. & Ishii, S. Inheritance of stress-induced, ATF-2-dependent epigenetic change. Cell 145, 1049–1061 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Carone, B. R. et al. Paternally induced transgenerational environmental reprogramming of metabolic gene expression in mammals. Cell 143, 1084–1096 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Ng, S. F. et al. Chronic high-fat diet in fathers programs β-cell dysfunction in female rat offspring. Nature 467, 963–966 (2010).

    Article  CAS  PubMed  Google Scholar 

  144. Kaati, G., Bygren, L. O. & Edvinsson, S. Cardiovascular and diabetes mortality determined by nutrition during parents' and grandparents' slow growth period. Eur. J. Hum. Genet. 10, 682–688 (2002).

    Article  CAS  PubMed  Google Scholar 

  145. Pembrey, M. E. et al. Sex-specific, male-line transgenerational responses in humans. Eur. J. Hum. Genet. 14, 159–166 (2006).

    Article  PubMed  Google Scholar 

  146. Rassoulzadegan, M. et al. RNA-mediated non-mendelian inheritance of an epigenetic change in the mouse. Nature 441, 469–474 (2006).

    Article  CAS  PubMed  Google Scholar 

  147. de Rooij, D. G. Spermatogonial stem cell renewal in the mouse. I. Normal situation. Cell Tissue Kinet. 6, 281–287 (1973).

    CAS  PubMed  Google Scholar 

  148. Oakberg, E. F. Spermatogonial stem-cell renewal in the mouse. Anat. Rec. 169, 515–531 (1971).

    Article  CAS  PubMed  Google Scholar 

  149. Zheng, K., Wu, X., Kaestner, K. H. & Wang, P. J. The pluripotency factor LIN28 marks undifferentiated spermatogonia in mouse. BMC Dev. Biol. 9, 38 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Nakagawa, T., Nabeshima, Y. & Yoshida, S. Functional identification of the actual and potential stem cell compartments in mouse spermatogenesis. Dev. Cell. 12, 195–206 (2007).

    Article  CAS  PubMed  Google Scholar 

  151. Lok, D. & de Rooij, D. G. Spermatogonial multiplication in the Chinese hamster. III. Labelling indices of undifferentiated spermatogonia throughout the cycle of the seminiferous epithelium. Cell Tissue Kinet. 16, 31–40 (1983).

    CAS  PubMed  Google Scholar 

  152. Nakagawa, T., Sharma, M., Nabeshima, Y., Braun, R. E. & Yoshida, S. Functional hierarchy and reversibility within the murine spermatogenic stem cell compartment. Science 328, 62–67 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Oatley, M. J., Kaucher, A. V., Racicot, K. E. & Oatley, J. M. Inhibitor of DNA binding 4 is expressed selectively by single spermatogonia in the male germline and regulates the self-renewal of spermatogonial stem cells in mice. Biol. Reprod. 85, 347–356 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Matson, C. K. et al. The mammalian doublesex homolog DMRT1 is a transcriptional gatekeeper that controls the mitosis versus meiosis decision in male germ cells. Dev. Cell. 19, 612–624 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Ballow, D., Meistrich, M. L., Matzuk, M. & Rajkovic, A. Sohlh1 is essential for spermatogonial differentiation. Dev. Biol. 294, 161–167 (2006).

    Article  CAS  PubMed  Google Scholar 

  156. Pangas, S. A. et al. Oogenesis requires germ cell-specific transcriptional regulators Sohlh1 and Lhx8. Proc. Natl Acad. Sci. USA 103, 8090–8095 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Toyoda, S. et al. Sohlh2 affects differentiation of KIT positive oocytes and spermatogonia. Dev. Biol. 325, 238–248 (2009).

    Article  CAS  PubMed  Google Scholar 

  158. Kuramochi-Miyagawa, S. et al. MVH in piRNA processing and gene silencing of retrotransposons. Genes Dev. 24, 887–892 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  159. Crittenden, S. L. et al. Regulation of the mitosis/meiosis decision in the Caenorhabditis elegans germline. Phil. Trans. R. Soc. Lond. B 358, 1359–1362 (2003).

    Article  CAS  Google Scholar 

  160. Sonoda, J. & Wharton, R. P. Recruitment of Nanos to hunchback mRNA by Pumilio. Genes Dev. 13, 2704–2712 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  161. Kim, J. Y., Lee, Y. C. & Kim, C. Direct inhibition of Pumilo activity by Bam and Bgcn in Drosophila germ line stem cell differentiation. J. Biol. Chem. 285, 4741–4746 (2010).

    Article  CAS  PubMed  Google Scholar 

  162. Xu, E. Y., Chang, R., Salmon, N. A. & Reijo Pera, R. A. A gene trap mutation of a murine homolog of the Drosophila stem cell factor Pumilio results in smaller testes but does not affect litter size or fertility. Mol. Reprod. Dev. 74, 912–921 (2007).

    Article  CAS  PubMed  Google Scholar 

  163. Chen, J. et al. Genome-wide analysis of translation reveals a critical role for deleted in azoospermia-like (Dazl) at the oocyte-to-zygote transition. Genes Dev. 25, 755–766 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Chuma, S. et al. Mouse Tudor Repeat-1 (MTR-1) is a novel component of chromatoid bodies/nuages in male germ cells and forms a complex with snRNPs. Mech. Dev. 120, 979–990 (2003).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors would like to acknowledge D. de Rooij for useful discussions during preparation of this review, and R. Desgraz, T. Endo, A. Godfrey, J. Hughes, M. Kojima, J. Mueller and K. Romer for comments on the manuscript.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Bluma J. Lesch.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

David C. Page's homepage

Confocal microscopy movies associated with reference 33, showing the migration of living, fluorescently labelled primordial germ cells in the mouse embryo

Glossary

Gametes

Haploid, differentiated germ cells: mature sperm and eggs.

Zygotic

Referring to a zygote, which is a one-celled embryo as the initial product of fertilization.

Germ plasm

Specialized cytoplasm that contains factors that are necessary and sufficient for germ cell specification. Germ plasm may or may not have a known physical correlate in a given species.

P granules

Cytoplasmic structures comprising the germ plasm in Caenorhabditis elegans.

Oocytes

Female germ cells that have initiated meiosis. Because meiosis is not complete in the oocyte until fertilization, mature female gametes are oocytes.

Polar granules

Cytoplasmic structures comprising the germ plasm in Drosophila melanogaster.

Epiblast

A cup-shaped sheet of cells derived from the inner cell mass that will eventually form all tissues of the embryo proper.

Extra-embryonic ectoderm

Ectodermal tissue that is derived from the epiblast but does not contribute to the embryo proper. Ectoderm is one of the three primary germ layers produced during early embryonic development.

Anterior visceral endoderm

(AVE). Cell layer underlying the epiblast in the mouse embryo. It does not contribute to the embryo proper but serves important signalling functions during embryogenesis.

Primordial germ cells

A term used for cells early in the germ cell lineage, before they have initiated meiosis or begun sex-specific differentiation.

Gastrulation

The process by which the three primitive germ layers are formed in the early embryo; it is one of the first major differentiation events in development.

Pre-initiation complex

A protein complex made up of general transcription factors that positions RNA polymerase II (RNAPII) at gene transcription start sites and positions DNA in the RNAPII active site.

Polycomb

A chromatin regulatory complex that represses gene expression; it is associated with deposition of the histone mark H3K27me3.

Germline stem cells

(GSCs). Proliferative cells that maintain germ cell production in the adult, often by dividing to produce one self-renewing and one differentiating daughter cell.

Niche

A microenvironment that promotes the maintenance of germline stem cells. The term may refer to the somatic cells that are responsible for creating this microenvironment, or to the physical location in which they reside.

Distal tip cell

A specialized somatic cell comprising the germ cell niche in Caenorhabditis elegans.

Spermatocytes

Male germ cells that have initiated meiosis.

Spermatogenesis

Refers to the entire process of sperm generation from mitotic precursor to mature sperm.

Oogenesis

The process of oocyte generation, from mitotic precursor cell to mature oocyte in meiotic arrest.

Eclosion

The transition from pupa to adult in insects: hatching from the pupal case.

Cap cells

Somatic cells that, together with terminal filament cells and escort cells, make up the germ cell niche in Drosophila melanogaster females. They directly contact germline stem cells and promote stem-cell maintenance.

Hub

The cone-shaped group of somatic cells comprising the germ cell niche in Drosophila melanogaster males.

Cystoblast

A germline stem cell daughter cell that has moved away from the niche and initiated differentiation.

Licensing

A process permitting primordial germ cells to respond to signals promoting meiosis and male or female differentiation.

Prophase

First phase of the meiotic or mitotic cell division (M phase), during which chromosomes condense. In meiosis, prophase occurs before meiosis I and is divided into the leptotene, zygotene, pachytene, diplotene, and diakinesis stages.

Residual body

An anucleate cytoplasmic structure remaining after budding of Caenorhabditis elegans spermatids.

H3.3

A histone H3 variant subunit that is associated with actively transcribed genes as well as with specific heterochromatic regions such as telomeres. Unlike H3.1 and H3.2, deposition is cell-cycle-independent.

Protamines

Highly basic, arginine-rich proteins that replace histones in packaging the genomes of haploid sperm. Packaging with protamines results in highly condensed genomic DNA.

Spermiogenesis

The process of differentiation in haploid sperm after meiosis has been completed, involving nuclear compaction, loss of cytoplasm and generation of a flagellum.

Leptotene

The first stage of meiotic prophase. Chromosomes begin to condense.

Zygotene

The second stage of meiotic prophase. Homologous chromosomes pair.

Pachytene

The third stage of meiotic prophase. Homologous chromosomes are tightly held together by the synaptonemal complex, and homologous recombination ('crossing over') begins.

Diplotene

The fourth stage of meiotic prophase. The synaptonemal complex breaks down, but homologous chromosomes are held together at sites of recombination.

Diakinesis

The final stage of meiotic prophase. Chromosomes condense further, the nuclear envelope breaks down and the meiotic spindle begins to form.

Pronucleus

A term for the nuclei of the male and female gametes after they have formed a single cell at fertilization, before the nuclei have fused.

Epiblast-like stem cells

(EpiSCs). Stem cells that are derived from the epiblast of postimplantation embryos; they exhibit a more restricted differentiation potential than naive embryonic stem cells.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lesch, B., Page, D. Genetics of germ cell development. Nat Rev Genet 13, 781–794 (2012). https://doi.org/10.1038/nrg3294

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrg3294

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing