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.

  • Article
  • Published:

Defective germline reprogramming rewires the spermatogonial transcriptome

Abstract

Defective germline reprogramming in Piwil4 (Miwi2)- and Dnmt3l-deficient mice results in the failure to reestablish transposon silencing, meiotic arrest and progressive loss of spermatogonia. Here we sought to understand the molecular basis for this spermatogonial dysfunction. Through a combination of imaging, conditional genetics and transcriptome analysis, we demonstrate that germ cell elimination in the respective mutants arises as a result of defective de novo genome methylation during reprogramming rather than because of a function for the respective factors within spermatogonia. In both Miwi2−/− and Dnmt3l−/− spermatogonia, the intracisternal-A particle (IAP) family of endogenous retroviruses is derepressed, but, in contrast to meiotic cells, DNA damage is not observed. Instead, we find that unmethylated IAP promoters rewire the spermatogonial transcriptome by driving expression of neighboring genes. Finally, spermatogonial numbers, proliferation and differentiation are altered in Miwi2−/− and Dnmt3l−/− mice. In summary, defective reprogramming deregulates the spermatogonial transcriptome and may underlie spermatogonial dysfunction.

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

Access options

Buy this article

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

Fig. 1: MIWI2 but not DNMT3L is expressed in adult mouse undifferentiated spermatogonia.
Fig. 2: MIWI2 is not required for homeostatic or regenerative spermatogenesis.
Fig. 3: Defective reprogramming affects undifferentiated spermatogonia numbers, proliferation and differentiation.
Fig. 4: Deregulation of IAP, but no DNA damage, is found in Miwi2/ and Dnmt3l/ undifferentiated spermatogonia.
Fig. 5: IAPs deregulate gene expression in Miwi2/ and Dnmt3l/ undifferentiated spermatogonia.
Fig. 6: Increase in de novo–identified weakly expressed transcripts in Miwi2- or Dnmt3l-deficient undifferentiated spermatogonia.
Fig. 7: Defective de novo DNA methylation is associated with deregulated gene expression in Miwi2/ and Dnmt3l−/− undifferentiated spermatogonia.

Similar content being viewed by others

References

  1. Chiquoine, A. D. The identification, origin, and migration of the primordial germ cells in the mouse embryo. Anat. Rec. 118, 135–146 (1954).

    Article  CAS  PubMed  Google Scholar 

  2. Ginsburg, M., Snow, M. H. & McLaren, A. Primordial germ cells in the mouse embryo during gastrulation. Development 110, 521–528 (1990).

    CAS  PubMed  Google Scholar 

  3. Lawson, K. A. & Hage, W. J. Clonal analysis of the origin of primordial germ cells in the mouse. Ciba Found. Symp. 182, 68–84 (1994).

    CAS  PubMed  Google Scholar 

  4. Kobayashi, T. et al. Principles of early human development and germ cell program from conserved model systems. Nature 546, 416–420 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Sasaki, K. et al. The germ cell fate of cynomolgus monkeys is specified in the nascent amnion. Dev. Cell 39, 169–185 (2016).

    Article  CAS  PubMed  Google Scholar 

  6. Oestrup, O. et al. From zygote to implantation: morphological and molecular dynamics during embryo development in the pig. Zuchthygiene 44(Suppl. 3), 39–49 (2009).

    Article  PubMed  Google Scholar 

  7. Surani, M. A., Barton, S. C. & Norris, M. L. Development of reconstituted mouse eggs suggests imprinting of the genome during gametogenesis. Nature 308, 548–550 (1984).

    Article  CAS  PubMed  Google Scholar 

  8. McGrath, J. & Solter, D. Completion of mouse embryogenesis requires both the maternal and paternal genomes. Cell 37, 179–183 (1984).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  10. Kobayashi, H. et al. High-resolution DNA methylome analysis of primordial germ cells identifies gender-specific reprogramming in mice. Genome Res. 23, 616–627 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Tang, W. W. et al. A unique gene regulatory network resets the human germline epigenome for development. Cell 161, 1453–1467 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Seisenberger, S. et al. The dynamics of genome-wide DNA methylation reprogramming in mouse primordial germ cells. Mol. Cell 48, 849–862 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Molaro, A. et al. Two waves of de novo methylation during mouse germ cell development. Genes Dev. 28, 1544–1549 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Guibert, S., Forné, T. & Weber, M. Global profiling of DNA methylation erasure in mouse primordial germ cells. Genome Res. 22, 633–641 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lane, N. et al. Resistance of IAPs to methylation reprogramming may provide a mechanism for epigenetic inheritance in the mouse. Genesis 35, 88–93 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Lees-Murdock, D. J., De Felici, M. & Walsh, C. P. Methylation dynamics of repetitive DNA elements in the mouse germ cell lineage. Genomics 82, 230–237 (2003).

    Article  CAS  PubMed  Google Scholar 

  17. Bourc’his, D., Xu, G. L., Lin, C. S., Bollman, B. & Bestor, T. H. Dnmt3L and the establishment of maternal genomic imprints. Science 294, 2536–2539 (2001).

    Article  PubMed  Google Scholar 

  18. Chedin, F., Lieber, M. R. & Hsieh, C. L. The DNA methyltransferase-like protein DNMT3L stimulates de novo methylation by Dnmt3a. Proc. Natl. Acad. Sci. USA 99, 16916–16921 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Suetake, I., Shinozaki, F., Miyagawa, J., Takeshima, H. & Tajima, S. DNMT3L stimulates the DNA methylation activity of Dnmt3a and Dnmt3b through a direct interaction. J. Biol. Chem. 279, 27816–27823 (2004).

    Article  CAS  PubMed  Google Scholar 

  20. Gowher, H., Liebert, K., Hermann, A., Xu, G. & Jeltsch, A. Mechanism of stimulation of catalytic activity of Dnmt3A and Dnmt3B DNA-(cytosine-C5)-methyltransferases by Dnmt3L. J. Biol. Chem. 280, 13341–13348 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Ooi, S. K. et al. DNMT3L connects unmethylated lysine 4 of histone H3 to de novo methylation of DNA. Nature 448, 714–717 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Bourc’his, D. & Bestor, T. H. Meiotic catastrophe and retrotransposon reactivation in male germ cells lacking Dnmt3L. Nature 431, 96–99 (2004).

    Article  PubMed  Google Scholar 

  23. Hata, K., Okano, M., Lei, H. & Li, E. Dnmt3L cooperates with the Dnmt3 family of de novo DNA methyltransferases to establish maternal imprints in mice. Development 129, 1983–1993 (2002).

    CAS  PubMed  Google Scholar 

  24. Barau, J. et al. The DNA methyltransferase DNMT3C protects male germ cells from transposon activity. Science 354, 909–912 (2016).

    Article  CAS  PubMed  Google Scholar 

  25. Aravin, A. A. et al. A piRNA pathway primed by individual transposons is linked to de novo DNA methylation in mice. Mol. Cell 31, 785–799 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Kuramochi-Miyagawa, S. et al. DNA methylation of retrotransposon genes is regulated by Piwi family members MILI and MIWI2 in murine fetal testes. Genes Dev. 22, 908–917 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. De Fazio, S. et al. The endonuclease activity of Mili fuels piRNA amplification that silences LINE1 elements. Nature 480, 259–263 (2011).

    Article  PubMed  Google Scholar 

  28. Kafri, T. et al. Developmental pattern of gene-specific DNA methylation in the mouse embryo and germ line. Genes Dev. 6, 705–714 (1992).

    Article  CAS  PubMed  Google Scholar 

  29. Li, J. Y., Lees-Murdock, D. J., Xu, G. L. & Walsh, C. P. Timing of establishment of paternal methylation imprints in the mouse. Genomics 84, 952–960 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Kato, Y. et al. Role of the Dnmt3 family in de novo methylation of imprinted and repetitive sequences during male germ cell development in the mouse. Hum. Mol. Genet. 16, 2272–2280 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Webster, K. E. et al. Meiotic and epigenetic defects in Dnmt3L-knockout mouse spermatogenesis. Proc. Natl. Acad. Sci. USA 102, 4068–4073 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Carmell, M. A. et al. MIWI2 is essential for spermatogenesis and repression of transposons in the mouse male germline. Dev. Cell 12, 503–514 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Hata, K., Kusumi, M., Yokomine, T., Li, E. & Sasaki, H. Meiotic and epigenetic aberrations in Dnmt3L-deficient male germ cells. Mol. Reprod. Dev. 73, 116–122 (2006).

    Article  CAS  PubMed  Google Scholar 

  34. La Salle, S. et al. Windows for sex-specific methylation marked by DNA methyltransferase expression profiles in mouse germ cells. Dev. Biol. 268, 403–415 (2004).

    Article  PubMed  Google Scholar 

  35. Sakai, Y., Suetake, I., Shinozaki, F., Yamashina, S. & Tajima, S. Co-expression of de novo DNA methyltransferases Dnmt3a2 and Dnmt3L in gonocytes of mouse embryos. Gene Expr. Patterns 5, 231–237 (2004).

    Article  CAS  PubMed  Google Scholar 

  36. La Salle, S. et al. Loss of spermatogonia and wide-spread DNA methylation defects in newborn male mice deficient in DNMT3L. BMC Dev. Biol. 7, 104 (2007).

    Article  PubMed  PubMed Central  Google Scholar 

  37. Shovlin, T. C. et al. Sex-specific promoters regulate Dnmt3L expression in mouse germ cells. Hum. Reprod. 22, 457–467 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Liao, H. F. et al. DNMT3L promotes quiescence in postnatal spermatogonial progenitor cells. Development 141, 2402–2413 (2014).

    Article  CAS  PubMed  Google Scholar 

  39. Carrieri, C. et al. A transit-amplifying population underpins the efficient regenerative capacity of the testis. J. Exp. Med. 214, 1631–1641 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Vasiliauskaitė, L. et al. A MILI-independent piRNA biogenesis pathway empowers partial germline reprogramming. Nat. Struct. Mol. Biol. 24, 604–606 (2017).

    Article  PubMed  Google Scholar 

  41. Buaas, F. W. et al. Plzf is required in adult male germ cells for stem cell self-renewal. Nat. Genet. 36, 647–652 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Yoshinaga, K. et al. Role of c-kit in mouse spermatogenesis: identification of spermatogonia as a specific site of c-kit expression and function. Development 113, 689–699 (1991).

    CAS  PubMed  Google Scholar 

  43. Bao, J. et al. Conditional inactivation of Miwi2 reveals that MIWI2 is only essential for prospermatogonial development in mice. Cell Death Differ. 21, 783–796 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Sadate-Ngatchou, P. I., Payne, C. J., Dearth, A. T. & Braun, R. E. Cre recombinase activity specific to postnatal, premeiotic male germ cells in transgenic mice. Genesis 46, 738–742 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Di Giacomo, M. et al. Multiple epigenetic mechanisms and the piRNA pathway enforce LINE1 silencing during adult spermatogenesis. Mol. Cell 50, 601–608 (2013).

    Article  PubMed  Google Scholar 

  46. Hobbs, R. M. et al. Functional antagonism between Sall4 and Plzf defines germline progenitors. Cell Stem Cell 10, 284–298 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Badea, T. C., Wang, Y. & Nathans, J. A noninvasive genetic/pharmacologic strategy for visualizing cell morphology and clonal relationships in the mouse. J. Neurosci. 23, 2314–2322 (2003).

    Article  CAS  PubMed  Google Scholar 

  48. Bucci, L. R. & Meistrich, M. L. Effects of busulfan on murine spermatogenesis: cytotoxicity, sterility, sperm abnormalities, and dominant lethal mutations. Mutat. Res. 176, 259–268 (1987).

    Article  CAS  PubMed  Google Scholar 

  49. 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 

  50. 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 

  51. Hara, K. et al. Mouse spermatogenic stem cells continually interconvert between equipotent singly isolated and syncytial states. Cell Stem Cell 14, 658–672 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Di Giacomo, M., Comazzetto, S., Sampath, S. C., Sampath, S. C. & O’Carroll, D. G9a co-suppresses LINE1 elements in spermatogonia. Epigenetics Chromatin 7, 24 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  53. Grabherr, M. G. et al. Full-length transcriptome assembly from RNA-Seq data without a reference genome. Nat. Biotechnol. 29, 644–652 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Inoue, K., Ichiyanagi, K., Fukuda, K., Glinka, M. & Sasaki, H. Switching of dominant retrotransposon silencing strategies from posttranscriptional to transcriptional mechanisms during male germ-cell development in mice. PLoS Genet. 13, e1006926 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Davis, M. P. et al. Transposon-driven transcription is a conserved feature of vertebrate spermatogenesis and transcript evolution. EMBO Rep. 18, 1231–1247 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kigami, D., Minami, N., Takayama, H. & Imai, H. MuERV-L is one of the earliest transcribed genes in mouse one-cell embryos. Biol. Reprod. 68, 651–654 (2003).

    Article  CAS  PubMed  Google Scholar 

  57. Peaston, A. E. et al. Retrotransposons regulate host genes in mouse oocytes and preimplantation embryos. Dev. Cell 7, 597–606 (2004).

    Article  CAS  PubMed  Google Scholar 

  58. Macfarlan, T. S. et al. Embryonic stem cell potency fluctuates with endogenous retrovirus activity. Nature 487, 57–63 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Göke, J. et al. Dynamic transcription of distinct classes of endogenous retroviral elements marks specific populations of early human embryonic cells. Cell Stem Cell 16, 135–141 (2015).

    Article  PubMed  Google Scholar 

  60. Chuong, E. B., Rumi, M. A., Soares, M. J. & Baker, J. C. Endogenous retroviruses function as species-specific enhancer elements in the placenta. Nat. Genet. 45, 325–329 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Franke, V. et al. Long terminal repeats power evolution of genes and gene expression programs in mammalian oocytes and zygotes. Genome Res. 27, 1384–1394 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Smith, Z. D. et al. A unique regulatory phase of DNA methylation in the early mammalian embryo. Nature 484, 339–344 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Okamoto, Y. et al. DNA methylation dynamics in mouse preimplantation embryos revealed by mass spectrometry. Sci. Rep. 6, 19134 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Oda, M., Oxley, D., Dean, W. & Reik, W. Regulation of lineage specific DNA hypomethylation in mouse trophectoderm. PLoS One 8, e68846 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Schroeder, D. I. et al. Early developmental and evolutionary origins of gene body DNA methylation patterns in mammalian placentas. PLoS Genet. 11, e1005442 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  66. Macfarlan, T. S. et al. Endogenous retroviruses and neighboring genes are coordinately repressed by LSD1/KDM1A. Genes Dev. 25, 594–607 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Matsui, T. et al. Proviral silencing in embryonic stem cells requires the histone methyltransferase ESET. Nature 464, 927–931 (2010).

    Article  CAS  PubMed  Google Scholar 

  68. Rowe, H. M. et al. KAP1 controls endogenous retroviruses in embryonic stem cells. Nature 463, 237–240 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Cammas, F. et al. Mice lacking the transcriptional corepressor TIF1β are defective in early postimplantation development. Development 127, 2955–2963 (2000).

    CAS  PubMed  Google Scholar 

  70. Dodge, J. E., Kang, Y. K., Beppu, H., Lei, H. & Li, E. Histone H3-K9 methyltransferase ESET is essential for early development. Mol. Cell. Biol. 24, 2478–2486 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Rowe, H. M. et al. TRIM28 repression of retrotransposon-based enhancers is necessary to preserve transcriptional dynamics in embryonic stem cells. Genome Res. 23, 452–461 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Karimi, M. M. et al. DNA methylation and SETDB1/H3K9me3 regulate predominantly distinct sets of genes, retroelements, and chimeric transcripts in mESCs. Cell Stem Cell 8, 676–687 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Hu, G. et al. A genome-wide RNAi screen identifies a new transcriptional module required for self-renewal. Genes Dev. 23, 837–848 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Urbán, N. et al. Return to quiescence of mouse neural stem cells by degradation of a proactivation protein. Science 353, 292–295 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Kim, E. et al. Rb family proteins enforce the homeostasis of quiescent hematopoietic stem cells by repressing Socs3 expression. J. Exp. Med. 214, 1901–1912 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Walter, D. et al. Exit from dormancy provokes DNA-damage-induced attrition in haematopoietic stem cells. Nature 520, 549–552 (2015).

    Article  PubMed  Google Scholar 

  77. Lay, K., Kume, T. & Fuchs, E. FOXC1 maintains the hair follicle stem cell niche and governs stem cell quiescence to preserve long-term tissue-regenerating potential. Proc. Natl. Acad. Sci. USA 113, E1506–E1515 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Uesaka, T. et al. Conditional ablation of GFRα1 in postmigratory enteric neurons triggers unconventional neuronal death in the colon and causes a Hirschsprung’s disease phenotype. Development 134, 2171–2181 (2007).

    Article  CAS  PubMed  Google Scholar 

  79. Yang, H. et al. One-step generation of mice carrying reporter and conditional alleles by CRISPR/Cas-mediated genome engineering. Cell 154, 1370–1379 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Morgan, M. et al. mRNA 3′ uridylation and poly(A) tail length sculpt the mammalian maternal transcriptome. Nature 548, 347–351 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The research leading to these results has received funding from the European Research Council under the European Union’s Seventh Framework Programme (FP7/2007-2013)–ERC grant agreement GA 310206. This study was technically supported by the EMBL Genomic Core facility, as well as EMBL Monterotondo’s genome engineering, flow cytometry and microscopy core facilities.

Author information

Authors and Affiliations

Authors

Contributions

L.V. contributed to the design, execution and analysis of the majority of experiments. R.V.B. generated the whole-genome bisulfite libraries and performed the bioinformatic analysis of genomic methylation under the supervision of W.R. L.V. and I.I. performed the in vivo characterization of spermatogonia. L.V. and C.C. designed, generated and validated the Dnmt3lV5 allele. A.J.E. performed all gene expression and transposon bioinformatic analyses. D.O’C. conceived and supervised this study. L.V. and D.O’C. wrote the final version of the manuscript.

Corresponding author

Correspondence to Dónal O’Carroll.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Generation and validation of the Dnmt3lV5 allele.

a, Targeting strategy for generation of the Dnmt3lV5 allele by CRISPR–Cas9 gene-editing technology. b, Validation of the Dnmt3lV5 allele by PCR using primers flanking the initial ATG codon. In the presence of the wild-type allele, a 411-bp fragment is amplified; in the presence of the Dnmt3lV5 allele, a 459-bp fragment is amplified. c, Normal testicular weight of adult Dnmt3lV5/+ and Dnmt3lV5/V5 mice. Error bars represent s.d. of the mean (n = 6 animals for Dnmt3lV5/+ and 5 animals for Dnmt3lV5/V5). Individual data points are shown. d, Representative images of hematoxylin- and eosin-stained testis cross-sections of wild-type and Dnmt3lV5/V5 mice. e, Representative images of wild-type and Dnmt3lV5/+ E16.5 fetal testis cross-sections stained with antibody against V5. f, Miwi2 and Dnmt3l mRNA expression levels in juvenile P14 undifferentiated spermatogonia as determined by RNA-seq. Source data for c are provided online.

Source data

Supplementary Figure 2 MIWI2 is not required for homeostatic or regenerative spermatogenesis.

a, Conversion of the Miwi2FL allele to the Miwi2 (Miwi2null) allele in Miwi2iKO mice after tamoxifen administration. b, Expression of Miwi2 transcript in Miwi2CTL and Miwi2iKO mice 2 weeks after the last tamoxifen injection. After tamoxifen administration, Miwi2iKO mice express Miwi2 transcript without exon 17, which was flanked by two loxP sites. Here primers spanning exons 16 and 17 were used for RT–qPCR analysis. Error bars represent s.d. of the mean (n = 3 animals). c, Scheme representing a timeline for inducible Miwi2 deletion (tamoxifen (Tmx) administration is depicted as black arrows) and the analysis time point (red bar) 2 weeks after the last tamoxifen injection. d, Representative images of hematoxylin- and eosin-stained testis cross-sections of tamoxifen-treated Miwi2CTL and Miwi2iKO mice. Respective time points after tamoxifen administration are shown. e, Representative images of hematoxylin- and eosin-stained testis cross-sections of tamoxifen- and busulfan-treated Miwi2CTL and Miwi2iKO mice. Respective time points after busulfan administration are shown.

Supplementary Figure 3 Generation and validation of the Dnmt3l allele.

a, Targeting strategy for generation of the Dnmt3l allele by homologous recombination. b, Southern blotting of the fragments generated by the BamHI restriction enzyme from wild-type (WT), Dnmt3lNeo/+ and Dnmt3l+/– genomic DNA hybridized with the external 3′ probe. c, Testicular weight of adult Dnmt3l+/– and Dnmt3l–/– mice. Error bars represent s.d. of the mean (n = 5 animals for Dnmt3l+/– and 7 animals for Dnmt3l–/–). Individual data points are shown. d, Representative images of hematoxylin- and eosin-stained testis cross-sections of WT and Dnmt3l–/– mice. e, Representative images of adult WT and Dnmt3l–/– testis cross-sections stained with antibody against LINE1 ORF1p. Source data for c are provided online.

Source data

Supplementary Figure 4 CD45CD51c-KitCD9+ surface expression identifies undifferentiated spermatogonia.

FACS analysis of testicular cell suspensions from adult Miwi2tdTom/+; Gfrα1GFP/+ mice. Representative example of a gating strategy for analyzing undifferentiated spermatogonia in adult Miwi2tdTom/+; Gfrα1GFP/+ mice, where a single-cell suspension of testicular cells was stained with CD45-biotin, CD51-biotin, streptavidin-qDot605, c-Kit-PE-Cy7 and CD9-APC antibodies. Only CD45CD51c-KitCD9+ cells express Miwi2-tdTom, Gfrα1-GFP or both Miwi2-tdTom and Gfrα1-GFP; thus, this surface stain identifies undifferentiated spermatogonia.

Supplementary Figure 5 Representative example of a gating strategy for sorting and/or analyzing Miwi2-tdTomato-expressing undifferentiated spermatogonia.

a, A single-cell suspension of testicular cells was stained with CD45-biotin, CD51-biotin, streptavidin-qDot605, c-Kit-PE-Cy7 and CD9-FITC antibodies to isolate and analyze CD45CD51c-KitMiwi2-tdTomato+CD9+ or analyze CD45CD51c-KitCD9+ cell populations. b, A single-cell suspension of testicular cells was stained with CD45-biotin, CD51-biotin, streptavidin-APC and c-Kit-PE-Cy7 antibodies and Hoechst to determine the DNA content of CD45CD51c-KitMiwi2-tdTomato+ cell populations.

Supplementary Figure 6 Impact of Miwi2 and Dnmt3l deficiency on juvenile P14 spermatogonia.

a, Total testicular cell number from Miwi2tdTom/+ (WT), Miwi2tdTom/tdTom (Miwi2–/–) and Dnmt3l–/–; Miwi2tdTom/+ (Dnmt3l–/–) juvenile P14 mice. Error bars represent s.d. of the mean (n = 8 animals for WT and 7 animals for Miwi2–/– and Dnmt3l–/–). Individual data points are shown. b, FACS analysis of live CD45CD51 gated testicular cells from juvenile P14 mice of the indicated genotypes. Numbers indicate the percentage of live CD45CD51c-KitCD9+ cells. c, Enumeration of live CD45CD51c-KitCD9+ cells per testis from juvenile P14 mice of the indicated genotypes (as defined in a). Error bars represent s.d. of the mean. Individual data points are shown. d, FACS analysis of live CD45CD51 gated testicular cells from juvenile P14 mice of the indicated genotypes. Numbers indicate the percentage of live CD45CD51c-KitMiwi2-tdTomato+ and CD45CD51c-Kit+Miwi2-tdTomato+ cells. e,f, Enumeration of live CD45CD51c-KitMiwi2-tdTomato+ (e) and live CD45CD51c-Kit+Miwi2-tdTomato+ (f) cells per testis from juvenile P14 mice of the indicated genotypes (as defined in a). Error bars represent s.d. of the mean. Individual data points are shown. The number of animals (n) in c, e and f is 8 for WT and Dnmt3l–/– and 7 for Miwi2–/–, except that 6 animals were used for Dnmt3l–/– in f. Significance in a, c, e and f was assessed using two-sided t test. n.s., not significant; *P < 0.01, **P < 0.001, ***P < 0.0001. Source data for a, c, e and f are provided online.

Source data

Supplementary Figure 7 Methylation changes in Miwi2–/– and Dnmt3l–/– juvenile P14 undifferentiated spermatogonia.

a, Comparison of the percentage of CpG methylation in WT and Miwi2–/– as well as WT and Dnmt3l–/– undifferentiated spermatogonia. Blue dots represent significant DMRs. Analysis was performed in biological triplicate (n = 3 animals). b,c, Percentages of CpG methylation levels measured by WGBS of WT (gray), Miwi2–/– (blue) and Dnmt3l–/– (green) undifferentiated spermatogonia for the indicated genomic features or TEs. An averaged value from biological triplicates (n = 3 animals) for each genotype is shown. The middle line represents the median of the data points with whiskers indicating the data range.

Supplementary information

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Vasiliauskaitė, L., Berrens, R.V., Ivanova, I. et al. Defective germline reprogramming rewires the spermatogonial transcriptome. Nat Struct Mol Biol 25, 394–404 (2018). https://doi.org/10.1038/s41594-018-0058-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-018-0058-0

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