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.

Co-repressor CBFA2T2 regulates pluripotency and germline development

Abstract

Developmental specification of germ cells lies at the heart of inheritance, as germ cells contain all of the genetic and epigenetic information transmitted between generations. The critical developmental event distinguishing germline from somatic lineages is the differentiation of primordial germ cells (PGCs)1,2, precursors of sex-specific gametes that produce an entire organism upon fertilization. Germ cells toggle between uni- and pluripotent states as they exhibit their own ‘latent’ form of pluripotency. For example, PGCs express a number of transcription factors in common with embryonic stem (ES) cells, including OCT4 (encoded by Pou5f1), SOX2, NANOG and PRDM14 (refs 2, 3, 4). A biochemical mechanism by which these transcription factors converge on chromatin to produce the dramatic rearrangements underlying ES-cell- and PGC-specific transcriptional programs remains poorly understood. Here we identify a novel co-repressor protein, CBFA2T2, that regulates pluripotency and germline specification in mice. Cbfa2t2−/− mice display severe defects in PGC maturation and epigenetic reprogramming. CBFA2T2 forms a biochemical complex with PRDM14, a germline-specific transcription factor. Mechanistically, CBFA2T2 oligomerizes to form a scaffold upon which PRDM14 and OCT4 are stabilized on chromatin. Thus, in contrast to the traditional ‘passenger’ role of a co-repressor, CBFA2T2 functions synergistically with transcription factors at the crossroads of the fundamental developmental plasticity between uni- and pluripotency.

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: PRDM14 and the co-repressor protein CBFA2T2 interact and bind to chromatin interdependently.
Figure 2: PRDM14 and CBFA2T2 regulate pluripotency.
Figure 3: Cbfa2t2−/− mice are defective in their germ line.
Figure 4: Mechanism of CBFA2T2–PRDM14 complex chromatin binding and direct regulation of PGC epigenetic reprogramming.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

Sequencing data have been deposited in the Gene Expression Omnibus under accession number GSE71676.

References

  1. 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  ADS  PubMed  Google Scholar 

  2. Surani, M. A., Hayashi, K. & Hajkova, P. Genetic and epigenetic regulators of pluripotency. Cell 128, 747–762 (2007)

    Article  CAS  PubMed  Google Scholar 

  3. Yeom, Y. I. et al. Germline regulatory element of Oct-4 specific for the totipotent cycle of embryonal cells. Development 122, 881–894 (1996)

    CAS  PubMed  Google Scholar 

  4. 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 

  5. Chia, N. Y. et al. A genome-wide RNAi screen reveals determinants of human embryonic stem cell identity. Nature 468, 316–320 (2010)

    Article  CAS  ADS  PubMed  Google Scholar 

  6. Ma, Z., Swigut, T., Valouev, A., Rada-Iglesias, A. & Wysocka, J. Sequence-specific regulator Prdm14 safeguards mouse ESCs from entering extraembryonic endoderm fates. Nature Struct. Mol. Biol . 18, 120–127 (2011)

    Article  CAS  Google Scholar 

  7. Yamaji, M. et al. PRDM14 ensures naive pluripotency through dual regulation of signaling and epigenetic pathways in mouse embryonic stem cells. Cell Stem Cell 12, 368–382 (2013)

    Article  CAS  PubMed  Google Scholar 

  8. Chan, Y. S. et al. A PRC2-dependent repressive role of PRDM14 in human embryonic stem cells and induced pluripotent stem cell reprogramming. Stem Cells 31, 682–692 (2013)

    Article  CAS  PubMed  Google Scholar 

  9. Payer, B. et al. Tsix RNA and the germline factor, PRDM14, link X reactivation and stem cell reprogramming. Mol. Cell 52, 805–818 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. De Braekeleer, E. et al. RUNX1 translocations and fusion genes in malignant hemopathies. Future Oncol. 7, 77–91 (2011)

    Article  CAS  PubMed  Google Scholar 

  11. Guastadisegni, M. C. et al. CBFA2T2 and C20orf112: two novel fusion partners of RUNX1 in acute myeloid leukemia. Leukemia 24, 1516–1519 (2010)

    Article  CAS  PubMed  Google Scholar 

  12. Miyoshi, H. et al. The t(8;21) translocation in acute myeloid leukemia results in production of an AML1-MTG8 fusion transcript. EMBO J. 12, 2715–2721 (1993)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Calabi, F. & Cilli, V. CBFA2T1, a gene rearranged in human leukemia, is a member of a multigene family. Genomics 52, 332–341 (1998)

    Article  CAS  PubMed  Google Scholar 

  14. Liu, Y. et al. The tetramer structure of the Nervy homology two domain, NHR2, is critical for AML1/ETO’s activity. Cancer Cell 9, 249–260 (2006)

    Article  PubMed  Google Scholar 

  15. Hug, B. A. & Lazar, M. A. ETO interacting proteins. Oncogene 23, 4270–4274 (2004)

    Article  CAS  PubMed  Google Scholar 

  16. Sun, X. J. et al. A stable transcription factor complex nucleated by oligomeric AML1-ETO controls leukaemogenesis. Nature 500, 93–97 (2013)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  17. Mikkelsen, T. S. et al. Dissecting direct reprogramming through integrative genomic analysis. Nature 454, 49–55 (2008)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  18. 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 

  19. Beard, C., Hochedlinger, K., Plath, K., Wutz, A. & Jaenisch, R. Efficient method to generate single-copy transgenic mice by site-specific integration in embryonic stem cells. Genesis 44, 23–28 (2006)

    Article  CAS  PubMed  Google Scholar 

  20. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nature Protocols 8, 2281–2308 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  22. Yang, H., Wang, H. & Jaenisch, R. Generating genetically modified mice using CRISPR/Cas-mediated genome engineering. Nature Protocols 9, 1956–1968 (2014)

    Article  CAS  PubMed  Google Scholar 

  23. Amann, J. M. et al. Mtgr1 is a transcriptional corepressor that is required for maintenance of the secretory cell lineage in the small intestine. Mol. Cell. Biol. 25, 9576–9585 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Whyte, W. A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Tachibana, M. et al. Histone methyltransferases G9a and GLP form heteromeric complexes and are both crucial for methylation of euchromatin at H3-K9. Genes Dev. 19, 815–826 (2005)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Seki, Y. et al. Extensive and orderly reprogramming of genome-wide chromatin modifications associated with specification and early development of germ cells in mice. Dev. Biol. 278, 440–458 (2005)

    Article  CAS  PubMed  Google Scholar 

  27. Hajkova, P. et al. Chromatin dynamics during epigenetic reprogramming in the mouse germ line. Nature 452, 877–881 (2008)

    Article  CAS  ADS  PubMed  Google Scholar 

  28. Nady, N. et al. ETO family protein Mtgr1 mediates Prdm14 functions in stem cell maintenance and primordial germ cell formation. Elife 4, e10150 (2015)

    Article  PubMed  PubMed Central  Google Scholar 

  29. Liew, C. G., Draper, J. S., Walsh, J., Moore, H. & Andrews, P. W. Transient and stable transgene expression in human embryonic stem cells. Stem Cells 25, 1521–1528 (2007)

    Article  CAS  PubMed  Google Scholar 

  30. Burton, A. et al. Single-cell profiling of epigenetic modifiers identifies PRDM14 as an inducer of cell fate in the mammalian embryo. Cell Reports 5, 687–701 (2013)

    Article  CAS  PubMed  Google Scholar 

  31. Kuzmichev, A., Jenuwein, T., Tempst, P. & Reinberg, D. Different EZH2-containing complexes target methylation of histone H1 or nucleosomal histone H3. Mol. Cell 14, 183–193 (2004)

    Article  CAS  PubMed  Google Scholar 

  32. Dignam, J. D., Lebovitz, R. M. & Roeder, R. G. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11, 1475–1489 (1983)

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Takahashi, Y., Rayman, J. B. & Dynlacht, B. D. Analysis of promoter binding by the E2F and pRB families in vivo: distinct E2F proteins mediate activation and repression. Genes Dev . 14, 804–816 (2000)

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Narendra, V. et al. CTCF establishes discrete functional chromatin domains at the Hox clusters during differentiation. Science 347, 1017–1021 (2015)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  35. Huang, W., Sherman, B. T. & Lempicki, R. A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature Protocols 4, 44–57 (2009)

    Article  CAS  Google Scholar 

  36. Wang, L., Feng, Z., Wang, X., Wang, X. & Zhang, X. DEGseq: an R package for identifying differentially expressed genes from RNA-seq data. Bioinformatics 26, 136–138 (2010)

    Article  PubMed  Google Scholar 

  37. Sommer, C. A. et al. Induced pluripotent stem cell generation using a single lentiviral stem cell cassette. Stem Cells 27, 543–549 (2009)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Mostoslavsky, G., Fabian, A. J., Rooney, S., Alt, F. W. & Mulligan, R. C. Complete correction of murine Artemis immunodeficiency by lentiviral vector-mediated gene transfer. Proc. Natl Acad. Sci. USA 103, 16406–16411 (2006)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  39. Onder, T. T. et al. Chromatin-modifying enzymes as modulators of reprogramming. Nature 483, 598–602 (2012)

    Article  CAS  ADS  PubMed  PubMed Central  Google Scholar 

  40. Lin, S. & Garcia, B. A. Examining histone posttranslational modification patterns by high-resolution mass spectrometry. Methods Enzymol. 512, 3–28 (2012)

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Yuan, Z. F. et al. EpiProfile quantifies histone peptides with modifications by extracting retention time and intensity in high-resolution mass spectra. Mol. Cell. Proteomics 14, 1696–1707 (2015)

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank P. Andrews and X.-J. Sun for providing plasmids. We are grateful to L. Vales, M. E. Torres-Padilla and L. Bu for critical comments, H. Zheng for MS analysis, and D. Hernandez, C. Leek, M. Yamaji, A. Paradkar and M. Alu for excellent technical assistance. The work was supported by the Howard Hughes Medical Institute (HHMI) and National Institutes of Health (NIH; RO1GM064844-12) (D.R.). B.A.G. acknowledges funding from NIH grant R01GM110174. T.T. was supported by the HHMI, NIH, Starr Foundation, and Tri-Institutional Stem Cell Initiative. M.Y. was a recipient of a Japan Society for the Promotion of Science (JSPS) Research Fellowship.

Author information

Authors and Affiliations

Authors

Contributions

S.T. designed and performed majority of experiments; V.N. analysed ChIP-seq and RNA-seq data; S.E.V., S.T. and M.S. performed iPSC reprogramming experiments; X.W. and B.A.G. quantified histone modifications. S.Y.K. did CRISPR zygotic injection. S.T., S.Y.K., M.Y. and L.A.R. characterized mice. M.Y., S.T. and T.T. designed and performed PGC experiments. S.T., V.N., M.Y. and D.R. wrote the manuscript.

Corresponding author

Correspondence to Danny Reinberg.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Extended data figures and tables

Extended Data Figure 1 Biochemical interaction between PRDM14 and CBFA2T2.

a, Mass spectrometry peptide counts from Flag affinity purification from NCCIT control cells and stable lines expressing Flag–HA–PRDM14 (PRDM14-F), and Flag–HA–CBFA2T2 (CBFA2T2-F). b, Characterization of in-house human PRDM14 antibody. Western blot performed using 30 μg of NCCIT and KH2 mES cell lysate. Human PRDM14 antibody is specific and does not cross-react with mouse PRDM14. c, Immunoprecipitation (IP) using antibodies against the indicated endogenous proteins in mES cells. d, Western blot of Superose 6 column fractionation of Flag-purified CBFA2T2 complex in NCCIT cells stably expressing Flag–HA–CBFA2T2. e, ChIP analysis using the indicated antibodies in 293T-REx harbouring a UAS-TK-Luciferase transgene. Fold enrichment represents the ratio of enrichment by ChIP-qPCR upon induction of GAL4–PRDM14 expression via doxycycline addition. Positions of the primer set are indicated by small arrows in the schematic. qPCR source data are included in the Supplementary Information.

Source data

Extended Data Figure 2 PRDM14 and CBFA2T2 exhibit an overlapping and interdependent distribution on chromatin in NCCIT cells.

a, Heat map depicting PRDM14, CBFA2T2, RING1B and SUZ12 read density across a 5-kb window centred about the PRDM14 (top) or RING1B (bottom) SERs identified in NCCIT cells. b, Representative genome browser tracks depicting SERs at the indicated genomic loci. c, Gene Ontology (GO) analysis of PRDM14 and CBFA2T2 common target genes. d, Western blot analysis of PRDM14 and CBFA2T2 protein levels in knockdown (KD) experiments (Fig. 1d, e).

Extended Data Figure 3 Characterization of knockout ES cell mutants and quantification of human iPS cell reprogramming efficiency.

a, b, Strategy for generating Prdm14- and Cbfa2t2-knockout (KO) mES cells via CRISPR–Cas9 genome editing. Sequencing chromatograms confirming homozygous disruption of the locus are depicted. c, Cbfa2t2- and Prdm14-knockout ES cells require 2i to maintain growth. ES cell lines generated under FBS plus LIF plus 2i conditions were continuously cultured in FBS plus LIF plus 2i (top, middle), or switched to FBS plus LIF (bottom). Eight days after 2i withdrawal (FBS plus LIF), well-formed ES cell colonies were undetectable; instead, mutant ES cells appeared to be differentiated. Scale bar, 100 μm. d, Proliferation rates of wild-type (WT) and mutant knockout ES cells as described in c. Data were obtained from three biological replicates. Please note error bars shown in the plots. Owing to the logarithmic scale used here, some error bars are very small and might be invisible. e, RNA-seq MA plot (log ratio (M) versus mean average (A)) in the indicated ES cells. Data are representative of three biological replicate experiments for each line. Mean abundance is plotted on the x axis and enrichment (both in log2 scale) is plotted on the y axis. Genes depicted in red are differentially expressed with a FDR < 0.0001. f, Heat map showing relative expression of all differentially expressed genes as described in Fig. 2c. The only difference is now the heat map is centred on CBFA2T2 differentially expressed genes, rather than PRDM14 differentially expressed genes. g, Scheme of human fibroblast reprogramming to iPS cells. Fibroblasts were transduced with lentiviruses expressing polycistronic OCT4/KLF4/SOX2/c-MYC (OKSM) and either PRDM14 or CBCFA2T2. Three weeks later, bright-field images of successfully reprogrammed colonies (left) and live TRA-1-81 staining (right) were recorded. Scale bar, 500 μm. h, Quantification of human iPS cell reprogramming efficiency based on TRA-1-81 staining with secondary antibody conjugated with horseradish peroxidase (HRP) and substrate DAB. Error bars are based on four biological replicates of each condition. The source data are included in the Supplementary Information.

Source data

Extended Data Figure 4 Cbfa2t2−/− mouse genotypes and sperm defects.

a, One representative Cbfa2t2−/− mouse genotype wherein a 7-bp fragment is deleted. b, Testes of multiple wild-type (n = 4) and Cbfa2t2−/− (n = 4) male mice at 8 weeks old were dissected and weighed. c, Number of sperm in the epididymis of Cbfa2t2+/+ (n = 4) and Cbfa2t2−/− (n = 4) mice is shown with standard error of the mean. P value was determined by Student’s t-test. d, Near loss of gonocytes in Cbfa2t2-knockout mutant P0 testes by DDX4 (MVH) staining. Visualization of MVH-positive (red) gonocytes in Cbfa2t2+/− (top) or Cbfa2t2−/− (bottom) testis at P0 stage. The merged images with Hoechst (left; white) are shown on the right. Scale bars, 100 mm. e, Numbers of AP-2γ-positive PGCs in Cbfa2t2+/+ (black), Cbfa2t2+/− (grey) and Cbfa2t2−/−(red) embryos at the indicated embryonic stages. LS, late-streak stage; EB, MB, and LB, early-, mid-, and late-bud stage; EHF, early-head fold stage; 2 st., 2 somites stage. Student’s t-test: *P = 0.03, **P = 0.003. f, Numbers of AP-2γ-positive PGCs in Cbfa2t2+/+ (black), Cbfa2t2+/− (grey) and Cbfa2t2−/−(red) embryos at the indicated embryonic stages. 0B, zero-bud stage; LHF, late-head fold stage. g, Left, expression of SOX2 (red) in AP-2γ-positive (green) PGCs in Cbfa2t2+/+ (top) or Cbfa2t2−/− (bottom) embryo at mid-bud stage, E7.25, shown as z-projection images of posterial confocal sections. Arrow indicates a minor PGC with relatively normal activation of SOX2. Scale bar, 50 mm. Right, percentage of SOX2-positive (red) cells in AP-2γ-positive (green) PGCs in the indicated genotypes of Cbfa2t2 at E7.0–7.25 (zero- to mid-bud stage) are shown with statistical significance (Student’s t-test: *P = 0.0006, **P = 0.0001; Cbfa2t2+/+, n = 7; Cbfa2t2+/−, n = 5; Cbfa2t2−/−, n = 5).

Extended Data Figure 5 Cbfa2t2-m7 mutant characterization and the related mechanism.

a, Gene Ontology (GO) analysis of PRDM14 ChIP-seq target genes. PRDM14 target genes are enriched in histone methyltransferase activities by DAVID functional annotation tool analysis. b, Cbfa2t2-m7 mutant genotyping. The mutant 7 amino acids are in red and corresponding wild-type (WT) residues are highlighted in blue in the displayed protein sequences. c, Bright-field images of wild-type and Cbfa2t2-m7 mES cells. Scale bar, 100 μm. d, Western blot analysis of PRDM14, CBFA2T2 and OCT4 protein levels in Prdm14-knockout (KO), Cbfa2t2-knockout or m7 mutant ES cells under feeder-free FBS plus LIF plus 2i condition. Nonspecific bands are denoted with an asterisk. e, ChIP-qPCR using antibodies against PRDM14, CBFA2T2 or OCT4 at selected OCT4 target genes. Occupancy is compared between wild-type, Cbfa2t2-knockout and Cbfa2t2-m7 mES cells. ChIP-qPCR primer sequences are included in Supplementary Table 3. f, RT–qPCR quantification of Ehmt1 mRNA levels in wild-type and mutant lines. P values are 0.004 (**) and 0.0142 (*).The source data are included in the Supplementary Information. g, Mass spectrometry quantification of histone H3K9 modifications in wild-type and mutant lines. P values are 0.00956, 0.04248 (*). The source data are included in the Supplementary Information. h, i, Additional immunofluorescence analysis of H3K9me2 (red) of AP-2γ-positive (green; arrowheads) PGCs in Cbfa2t2+/− and Cbfa2t2−/− embryos at E8.75 as described in Fig. 4i.

Source data

Supplementary information

Supplementary Table 1

This table contains PRDM14 and CBFA2T2 target genes identified by ChIP-seq in NCCIT cell. (XLSX 155 kb)

Supplementary Table 2

This table contains differentially expressed genes in Prdm14 and Cbfa2t2 Knockout lines. (XLSX 361 kb)

Supplementary Table 3

This table contains qPCR and PCR primers used in ChIP-qPCR and off-target verification. (XLSX 43 kb)

Supplementary Table 4

This table contains PRDM14 and CBFA2T2 target genes identified by ChIP-seq in mouse ESC. (XLSX 182 kb)

Supplementary Information

This file contains Supplementary Figure 1, the uncropped western blot images. (PDF 367 kb)

PowerPoint slides

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Tu, S., Narendra, V., Yamaji, M. et al. Co-repressor CBFA2T2 regulates pluripotency and germline development. Nature 534, 387–390 (2016). https://doi.org/10.1038/nature18004

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature18004

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

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