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:

Generation of functional oocytes from male mice in vitro

Abstract

Sex chromosome disorders severely compromise gametogenesis in both males and females. In oogenesis, the presence of an additional Y chromosome or the loss of an X chromosome disturbs the robust production of oocytes1,2,3,4,5. Here we efficiently converted the XY chromosome set to XX without an additional Y chromosome in mouse pluripotent stem (PS) cells. In addition, this chromosomal alteration successfully eradicated trisomy 16, a model of Down’s syndrome, in PS cells. Artificially produced euploid XX PS cells differentiated into mature oocytes in culture with similar efficiency to native XX PS cells. Using this method, we differentiated induced pluripotent stem cells from the tail of a sexually mature male mouse into fully potent oocytes, which gave rise to offspring after fertilization. This study provides insights that could ameliorate infertility caused by sex chromosome or autosomal disorders, and opens the possibility of bipaternal reproduction.

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: Sex conversion in ES cells and oocyte production in vitro.
Fig. 2: Oocyte production from XY ES cells with autosomal aneuploidy.
Fig. 3: Pups from oocytes differentiated in vitro from sex-converted iPS cells.
Fig. 4: CD38 expression in XO and XX PS cells.

Similar content being viewed by others

Data availability

The DNA-seq data have been deposited at the Sequence Read Archive (SRA) database under accession number PRJNA766461 and the RNA-seq data have been deposited at the Gene Expression Omnibus (GEO) database under accession number GSE184771.  Source data are provided with this paper.

Code availability

Custom code used in this article can be accessed at https://github.com/kentamurakami1986/oocytes-from-male-mice.

References

  1. Vernet, N. et al. The expression of Y-linked Zfy2 in XY mouse oocytes leads to frequent meiosis 2 defects, a high incidence of subsequent early cleavage stage arrest and infertility. Development 141, 855–866 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Amleh, A., Ledee, N., Saeed, J. & Taketo, T. Competence of oocytes from the B6.YDOM sex-reversed female mouse for maturation, fertilization, and embryonic development in vitro. Dev. Biol. 178, 263–275 (1996).

    Article  CAS  PubMed  Google Scholar 

  3. Mahadevaiah, S. K., Lovell-Badge, R. & Burgoyne, P. S. Tdy-negative XY, XXY and XYY female mice: breeding data and synaptonemal complex analysis. J. Reprod. Fertil. 97, 151–160 (1993).

    Article  CAS  PubMed  Google Scholar 

  4. Taketo-Hosotani, T., Nishioka, Y., Nagamine, C. M., Villalpando, I. & Merchant-Larios, H. Development and fertility of ovaries in the B6.YDOM sex-reversed female mouse. Development 107, 95–105 (1989).

    Article  CAS  PubMed  Google Scholar 

  5. Lavery, R. et al. XY Sox9 embryonic loss-of-function mouse mutants show complete sex reversal and produce partially fertile XY oocytes. Dev. Biol. 354, 111–122 (2011).

    Article  CAS  PubMed  Google Scholar 

  6. Saitou, M. & Yamaji, M. Primordial germ cells in mice. Cold Spring Harb. Perspect. Biol. 4, a008375 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Adams, I. R. & McLaren, A. Sexually dimorphic development of mouse primordial germ cells: switching from oogenesis to spermatogenesis. Development 129, 1155–1164 (2002).

    Article  CAS  PubMed  Google Scholar 

  8. Koopman, P., Gubbay, J., Vivian, N., Goodfellow, P. & Lovell-Badge, R. Male development of chromosomally female mice transgenic for Sry. Nature 351, 117–121 (1991).

    Article  ADS  CAS  PubMed  Google Scholar 

  9. Taketo, T. The role of sex chromosomes in mammalian germ cell differentiation: can the germ cells carrying X and Y chromosomes differentiate into fertile oocytes? Asian J. Androl. 17, 360–366 (2015).

    Article  CAS  PubMed  Google Scholar 

  10. Taketo, T. & Naumova, A. K. Oocyte heterogeneity with respect to the meiotic silencing of unsynapsed X chromosomes in the XY female mouse. Chromosoma 122, 337–349 (2013).

    Article  PubMed  Google Scholar 

  11. Alton, M., Lau, M. P., Villemure, M. & Taketo, T. The behavior of the X- and Y-chromosomes in the oocyte during meiotic prophase in the B6.Y(TIR)sex-reversed mouse ovary. Reproduction 135, 241–252 (2008).

    Article  CAS  PubMed  Google Scholar 

  12. Hikabe, O. et al. Reconstitution in vitro of the entire cycle of the mouse female germ line. Nature 539, 299–303 (2016).

    Article  ADS  PubMed  Google Scholar 

  13. Hamada, N. et al. Germ cell-intrinsic effects of sex chromosomes on early oocyte differentiation in mice. PLoS Genet. 16, e1008676 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Yamamoto, S. et al. Rapid selection of XO embryonic stem cells using Y chromosome-linked GFP transgenic mice. Transgenic Res. 23, 757–765 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Deng, J. M. et al. Generation of viable male and female mice from two fathers. Biol. Reprod. 84, 613–618 (2011).

    Article  CAS  PubMed  Google Scholar 

  16. Eggan, K. et al. Male and female mice derived from the same embryonic stem cell clone by tetraploid embryo complementation. Nat. Biotechnol. 20, 455–459 (2002).

    Article  CAS  PubMed  Google Scholar 

  17. Rudd, M. K. et al. Monosomy X rescue explains discordant NIPT results and leads to uniparental isodisomy. Prenat. Diagn. 38, 920–923 (2018).

    Article  CAS  PubMed  Google Scholar 

  18. Luo, Y. et al. Uniparental disomy of the entire X chromosome in Turner syndrome patient-specific induced pluripotent stem cells. Cell Discov. 1, 15022 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Denes, A. M., Landin-Wilhelmsen, K., Wettergren, Y., Bryman, I. & Hanson, C. The proportion of diploid 46,XX cells increases with time in women with Turner syndrome-a 10-year follow-up study. Genet. Test. Mol. Biomarkers 19, 82–87 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bolton, H. et al. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat. Commun. 7, 11165 (2016).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  21. Nakai-Futatsugi, Y. & Niwa, H. Zscan4 is activated after telomere shortening in mouse embryonic stem cells. Stem Cell Rep. 6, 483–495 (2016).

    Article  CAS  Google Scholar 

  22. Hayashi, K. et al. Offspring from oocytes derived from in vitro primordial germ cell-like cells in mice. Science 338, 971–975 (2012).

    Article  ADS  CAS  PubMed  Google Scholar 

  23. Codner, G. F. et al. Aneuploidy screening of embryonic stem cell clones by metaphase karyotyping and droplet digital polymerase chain reaction. BMC Cell Biol. 17, 30 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Gaztelumendi, N. & Nogues, C. Chromosome instability in mouse embryonic stem cells. Sci Rep. 4, 5324 (2014).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  25. Roper, R. J., St John, H. K., Philip, J., Lawler, A. & Reeves, R. H. Perinatal loss of Ts65Dn Down syndrome mice. Genetics 172, 437–443 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  Google Scholar 

  27. Severance, A. L., Midic, U. & Latham, K. E. Genotypic divergence in mouse oocyte transcriptomes: possible pathways to hybrid vigor impacting fertility and embryogenesis. Physiol. Genomics 52, 96–109 (2020).

    Article  CAS  PubMed  Google Scholar 

  28. Xie, B. et al. Poly(ADP-ribose) mediates asymmetric division of mouse oocyte. Cell Res. 28, 462–475 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Zuo, E. et al. CRISPR/Cas9-mediated targeted chromosome elimination. Genome Biol. 18, 224 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Choi, J. et al. Prolonged Mek1/2 suppression impairs the developmental potential of embryonic stem cells. Nature 548, 219–223 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang, M. et al. Aneuploid embryonic stem cells exhibit impaired differentiation and increased neoplastic potential. EMBO J. 35, 2285–2300 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Hirota, T. et al. Fertile offspring from sterile sex chromosome trisomic mice. Science 357, 932–935 (2017).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  33. Liu, P. et al. Passage number is a major contributor to genomic structural variations in mouse iPSCs. Stem Cells 32, 2657–2667 (2014).

    Article  CAS  PubMed  Google Scholar 

  34. Young, M. A. et al. Background mutations in parental cells account for most of the genetic heterogeneity of induced pluripotent stem cells. Cell Stem Cell 10, 570–582 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Liang, Q., Conte, N., Skarnes, W. C. & Bradley, A. Extensive genomic copy number variation in embryonic stem cells. Proc. Natl Acad. Sci. USA 105, 17453–17456 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  36. Li, Z. K. et al. Generation of bimaternal and bipaternal mice from hypomethylated haploid ESCs with imprinting region deletions. Cell Stem Cell 23, 665–676.e664 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Vaz, B., El Mansouri, F., Liu, X. & Taketo, T. Premature ovarian insufficiency in the XO female mouse on the C57BL/6J genetic background. Mol. Hum. Reprod. 26, 678–688 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ishikura, Y. et al. In vitro derivation and propagation of spermatogonial stem cell activity from mouse pluripotent stem cells. Cell Rep. 17, 2789–2804 (2016).

    Article  CAS  PubMed  Google Scholar 

  39. Ohta, H. et al. In vitro expansion of mouse primordial germ cell-like cells recapitulates an epigenetic blank slate. EMBO J. 36, 1888–1907 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Ying, Q. L. et al. The ground state of embryonic stem cell self-renewal. Nature 453, 519–523 (2008).

    Article  ADS  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hayashi, K. & Saitou, M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat. Protoc. 8, 1513–1524 (2013).

    Article  CAS  PubMed  Google Scholar 

  42. D’Hulst, C., Parvanova, I., Tomoiaga, D., Sapar, M. L. & Feinstein, P. Fast quantitative real-time PCR-based screening for common chromosomal aneuploidies in mouse embryonic stem cells. Stem Cell Rep. 1, 350–359 (2013).

    Article  Google Scholar 

  43. Scheinin, I. et al. DNA copy number analysis of fresh and formalin-fixed specimens by shallow whole-genome sequencing with identification and exclusion of problematic regions in the genome assembly. Genome Res. 24, 2022–2032 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Li, H. & Durbin, R. Fast and accurate short read alignment with Burrows-Wheeler transform. Bioinformatics 25, 1754–1760 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Danecek, P. et al. Twelve years of SAMtools and BCFtools. Gigascience 10, giab008 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Hayashi, K., Hikabe, O., Obata, Y. & Hirao, Y. Reconstitution of mouse oogenesis in a dish from pluripotent stem cells. Nat. Protoc. 12, 1733–1744 (2017).

    Article  CAS  PubMed  Google Scholar 

  48. Kim, D., Paggi, J. M., Park, C., Bennett, C. & Salzberg, S. L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 37, 907–915 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    Article  CAS  PubMed  Google Scholar 

  50. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Sasaki, K. et al. Robust in vitro induction of human germ cell fate from pluripotent stem cells. Cell Stem Cell 17, 178–194 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Kitajima and C. Meno for providing microscopes; N. Konishi for technical support; H. Toh for advice about DNA-seq; and Research Support Center, Research Center for Human Disease Modeling, Kyushu University Graduate School of Medical Sciences for technical assistance. This study was supported in part by Grant-in-Aid for Scientific Research on Innovative Areas from JSPS (grants 18H05544 and 18H05545 to K.H. and 18H05549 to T.S.K.); by a Grant-in-Aid for Specially Promoted Research from JSPS (grants 17H06098 and 22H04920 to M.S.); by the Takeda Science Foundation (K.H.); by the Luca Bella Foundation (K.H.); and by a Grant-in-Aid from The Open Philanthropy Project (K.H.).

Author information

Authors and Affiliations

Authors

Contributions

K.H. conceived the project. K.M., N. Hamazaki and K.H. performed the investigation. N. Hamazaki, T.S.K. and I.O. contributed the methodology. K.M. performed the DNA-seq and RNA-seq analyses. N. Hamada, G.N., H.O., Y.N., Y.I., Y.S., Y.K., F.A., K.A. and M.S. provided the resources. K.K. and K.H. supervised the project. K.M. and K.H. wrote the manuscript, incorporating feedback from all the authors.

Corresponding author

Correspondence to Katsuhiko Hayashi.

Ethics declarations

Competing interests

K.H. is an inventor on patent applications relating to in vitro oocyte production in mice filed by Kyushu University.

Peer review

Peer review information

Nature thanks Diana Laird and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer review reports are available.

Additional information

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

Extended data figures and tables

Extended Data Fig. 1 Generation of XX ES cells from XO ES cells with reversine.

(a) Knock-in of DsRed into the X chromosome. The schematic diagram shows the gRNA position, primers for genotyping (arrows), and expected sizes of the amplicons. The right image shows the result of PCR using the primers. The black dots indicate the ES cell lines used in this study. M, size marker. W, wild type. (b) Removal of the Y chromosome. Shown are the results of PCR analysis of sex chromosomes. Each sex chromosome is indicated at the left side of the image. Black dots indicate XO ES cell clones. The lane with an asterisk was excluded from the count because the band was faint. (c) Effect of reversine on the proliferation of ES cells at each passage. The results of three independent experiments are shown as mean±s.e.m. (d) The effect of reversine treatment on the proliferation of ES cells at day 2. The results of three independent experiments are shown as mean±s.e.m. P value was determined by One-way ANOVA with Bonferroni adjustment. (e) Effect of reversine on DsRed intensity of XRO ES cells in the total cell population and G1 population. The top row shows the DsRed intensity of the total cell population cultured without (Control) or with reversine at each concentration. The numbers show the percentage of the cell population at each reversine concentration. The second row shows the overlay of the DsRed intensity of the total cell population cultured without (grey) and with reversine (red) at each concentration. The third row shows the overlay of the cell size measured by forward scatter (FSC) of the total cell population cultured without (grey) and with reversine (red) at each concentration. The fourth row shows the DNA content in the total cell population cultured without (Control) or with reversine at each concentration. The G1 population in each plot was analyzed in the fifth and bottom rows. The fifth row shows the overlay of the DsRed intensity of the G1 population cultured without (grey) and with reversine (red) at each concentration. The bottom row shows the overlay of the cell size measured by forward scatter (FSC) of the G1 population cultured without (grey) and with reversine (red) at each concentration. Note that DsRed-highly positive cells increased in proportion to the concentration of reversine in both the total cell and G1 populations, whereas the cell size became diverse in both the small and large fractions. (f) DNA-seq analysis of sex-converted ES cell clones. Shown are the results of sex-converted ES cell clones apart from the ES cell clone shown in Fig. 1e. Two copies are plotted at 0 in log2 ratio. The numbers below the plot are the chromosome numbers.

Source data

Extended Data Fig. 2 PGCL cell induction and transcriptome analysis of oocytes from the sex-converted XX ES cells.

(a) PGCL cell induction from XRY, XRO and XRXR ES cells. The numbers in the FACS analysis images are the percentages of PGCL cells. Scale bars, 200 µm. (b) H3K27me3 states indicative of X chromosome states during PGCL cell induction. Images show immunofluorescence analysis of ES cells and PGCL cells at day 3 of induction (d3 PGCL cells). Note that single H3K27me3 bright spots corresponding to Xi, were observed in cells at day 3 of PGCL cell induction. Scale bars, 10 μm. The right graph shows the percentage of cells with the indicated H3K27me3 state in BV positive and negative cells at day 3 of PGCL cell induction. These results are based on biologically duplicated samples. (c) Gene expression analysis of GV (left) and MII (right) oocytes differentiated from native XX ES cells (BVSCH18) and XRXR ES cells (BVSCN2-4-63R2). Genes expressed more than twofold higher (red) or twofold lower (blue) in XRXR oocytes are shown. The FDR cutoff is 0.01. Biological duplicate samples.

Source data

Extended Data Fig. 3 Reversine effect on trisomy 16 ES cells and oocyte production from trisomy 16 XY and XO ES cells.

(a) DNA-seq analysis of a mosaic trisomy 16 clone. (b) DNA-seq analysis of trisomy 16 XRO ES cells. Another trisomy 16 XRO cell line is shown in Fig. 2b. (c) Q-PCR analysis to determine the X chromosome number. Blue and orange dots show the X chromosome content determined using chromosome 7 and chromosome 11, respectively, as references. Details are described in Fig. 1c. The genotypes of sex chromosomes and individual clone IDs are shown below the graph. (d) DNA-seq analysis of FACS sorted trisomy 16 XRO ES cells without (control) or with reversine treatment. The left column shows the whole population, and the right column shows the top 5% fraction of the DsRed-high population. (e) Log2 ratio of autosomes and the X chromosome under each condition in the DNA-seq analysis. Note that the ratio of XRXR ES cells was significantly increased in the top 5% fraction compared to the total cell fraction with 0.02 µM reversine, while autosomes were relatively unaffected. There was no significant difference (P > 0.11) in the ratio of XRXR ES cells in the top 5% fraction without and with 0.02 µM reversine, consistent with the results shown in Fig. 1b. P value was determined by One-way ANOVA with Bonferroni adjustment. The values were compiled from biologically triplicated experiments. (f) PGCL cell induction from trisomy 16 XRY ES cells and trisomy 16 XRO ES cells. Shown are representative images and FACS analyses of the induction of PGCL cells from the ES cells indicated. The numbers in the FACS analysis are the percentages of PGCL cells. Scale bars, 200 µm. (g) IVDi culture using PGCL cells from trisomy 16 XRY ES cells and trisomy 16 XRO ES cells. Representative images of the rOvaries at the days indicated are shown. Scale bars, 200 µm. Similar results were obtained in biologically duplicated experiments.

Source data

Extended Data Fig. 4 Generation of XX iPS cells from XY iPS cells followed by oocyte production.

(a) Genotype of XY iPS cells used in this study. Shown are the results of PCR analysis of sex chromosomes (top) and knock-in of DsRed into the X chromosome (bottom). Details for the knock-in are shown in Extended Data Fig. 1a. All six iPS cell lines tested had X and Y chromosomes (top). The black dot in each image indicates the iPS cell clone used in this study. The knock-in alleles were detected in four of the six iPS cell lines. The same results were obtained in PCR experiment repeated twice. (b) Removal of the Y chromosome. Shown are the results of PCR analysis of sex chromosomes after 9 passages. The black dot indicates XRO iPS cells used in this study. The same results were obtained in PCR experiment repeated twice. (c) Q-PCR analysis to determine the X chromosome number. Blue and orange dots show the X chromosome content determined using chromosome 7 and chromosome 11, respectively, as references. Details are described in Fig. 1c. The genotypes of sex chromosomes and individual clone IDs are shown below the graph. (d) DNA-seq analysis of sex-converted iPS cell clones. Two copies are plotted at 0 in log2 ratio. The numbers below the plot are the chromosome numbers. The genotypes and the IDs of the individual clones are displayed on the left side of the plot. (e) PGCL cell induction from XRY, XRO and XRXR iPS cells. Shown are representative images and FACS analyses of the induction of PGCL cells from the iPS cells indicated. The numbers in the FACS analysis are the percentages of PGCL cells. Scale bars, 200 µm. Similar results were obtained in biologically duplicated experiments. (f) IVDi culture using PGCL cells from XRY, XRO and XRXR iPS cells. Shown are representative images of the rOvaries at the days indicated. As a negative control, a representative rOvary containing only embryonic gonadal somatic cells is shown. The images at day 21 of culture are the same as in Fig. 3b. Scale bars, 200 µm. Similar results were obtained in biologically duplicated experiments.

Source data

Extended Data Fig. 5 Derivation of offspring from a single iPS cell line.

(a) Chimeric mice harbouring XRY or sex-converted XRXR iPS cells. The graph below the images shows the ratio of iPS cell chimeric mice. The number in the top graph is the percentage of male chimeras harbouring XRY iPS cells and the numbers in the middle and bottom graphs are the percentages of female chimeras harbouring XRXR iPS cells. (b) Offspring from chimeric mice harbouring XRY or sex-converted XRXR iPS cells. The top images show offspring from male chimeric mice harbouring XRY iPS cells (left) and offspring female chimeric mice harbouring XRXR iPS cells (right). The middle graph shows the coat colour of offspring from a mating between chimeric mice and wild-type albino mice. The number in the graph is the percentage of non-albino mice from a mating between chimeric mice and wild type albino mice. The bottom gel image show genotyping of the offspring from iPS cell chimeric mice harbouring XRY iPS cells (left) or sex-converted XRXR iPS cells (right). PCR products from BV and DsRed transgenes, exogenous retroviral Sox2 and Pou5f1, and the Prdm 14 locus as a positive control are shown. The coat colour of number 10 was albino. P, positive control. N, negative control. Details of the positive control and negative control are described in Fig. 3d. (c) Pups from a mating between an XRY iPS cell-chimeric male and XRXR iPS cell-chimeric female mouse. (d) FACS analysis of H-2K haplotypes of offspring obtained from crosses between the chimeric mice. Shown are the FACS plot of H-2K haplotypes in lymphocytes of C57BL/6J (H-2Kb), 129X1/SvJ (H-2Kb), Balb/c (H-2Kd), and F1 mice from a mating between chimeric mice. Note that one of the F1 mice had only H-2Kb, whereas the other had a hybrid haplotype. (e) SNP analysis of offspring. Shown are SNPs at rs3022980 and rs3022981 in C57BL/6J, 129X1/SvJ, BALB/c and F1 mice from a mating between chimeric mice. (f) Ratio of homozygous F1 from a mating between an XRY iPS cell-chimeric mouse and XRXR iPS cell-chimeric mouse. (g) An adult mouse from (c) at 10 months after birth.

Source data

Extended Data Fig. 6 Generation of XX ES cells from XO ES cells using anti-CD38 antibodies.

(a) FACS analysis of CD38 expression in C57BL/6J (originally XX) ES cells. Note that there are two types of CD38 expression: low and high. (b) FACS analysis of DsRed and CD38 in XRO and XRXR ES cells. Note that the CD38 intensity was increased in XRXR ES cells in proportion to the intensity of DsRed. (c) The gates used to select XX ES cells from XO ES cells using anti-CD38 antibodies. The gate at left (solid line) was used for the CD38-high cell population and the gate at right (dashed line) was used for the CD38-extremely high cell population. (d) Q-PCR analysis to determine the X chromosome number. The left graph shows the results using BVSC XO ES cells with high CD38 (the solid gate in (c)) and the right graph shows the results using BVSC XO ES cells with extremely high CD38 (the dashed gate in (c)). (e) The graph shows the results using non-reporter XO ES cells with high CD38. The genotypes of sex chromosomes and individual clone IDs are shown below the graph. Blue and orange dots show the X chromosome content determined using chromosome 7 and chromosome 11, respectively, as references. Details are described in Fig. 1c. (f) FACS analysis of PGCL cells derived from non-reporter and X-duplicated ESCs. PGCL cells were identified by antibodies against SSEA1 and CD6126. Similar results were obtained in biologically duplicated experiments.

Source data

Extended Data Table 1 Frequency of Y-chromosome loss in XY ES cells and iPS cells
Extended Data Table 2 Percentage of MII oocytes after IVM
Extended Data Table 3 Percentage of 2-cell embryos after IVF using iPS93C-49R6
Extended Data Table 4 Birth rate of the 2-cell embryos transferred using iPS93C-49R6

Supplementary information

Supplementary Information

This file contains Supplementary Figs. 1–7 and Supplementary Tables 1 and 2.

Reporting Summary

Peer Review File

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Murakami, K., Hamazaki, N., Hamada, N. et al. Generation of functional oocytes from male mice in vitro. Nature 615, 900–906 (2023). https://doi.org/10.1038/s41586-023-05834-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41586-023-05834-x

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research