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.

  • Resource
  • Published:

Inducible histone K-to-M mutations are dynamic tools to probe the physiological role of site-specific histone methylation in vitro and in vivo

Abstract

Development and differentiation are associated with profound changes to histone modifications, yet their in vivo function remains incompletely understood. Here, we generated mouse models expressing inducible histone H3 lysine-to-methionine (K-to-M) mutants, which globally inhibit methylation at specific sites. Mice expressing H3K36M developed severe anaemia with arrested erythropoiesis, a marked haematopoietic stem cell defect, and rapid lethality. By contrast, mice expressing H3K9M survived up to a year and showed expansion of multipotent progenitors, aberrant lymphopoiesis and thrombocytosis. Additionally, some H3K9M mice succumbed to aggressive T cell leukaemia/lymphoma, while H3K36M mice exhibited differentiation defects in testis and intestine. Mechanistically, induction of either mutant reduced corresponding histone trimethylation patterns genome-wide and altered chromatin accessibility as well as gene expression landscapes. Strikingly, discontinuation of transgene expression largely restored differentiation programmes. Our work shows that individual chromatin modifications are required at several specific stages of differentiation and introduces powerful tools to interrogate their roles in vivo.

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: Doxycycline-inducible K-to-M histone mutants globally suppress site-specific histone methylation and impair differentiation of ES cells.
Fig. 2: In vivo expression of H3K36M leads to rapid death with severe anaemia and organ abnormalities.
Fig. 3: In vivo expression of H3K36M dramatically alters myeloid and lymphoid differentiation.
Fig. 4: Expression of H3K36M and H3K9M have opposing, cell autonomous effects on HSPC function.
Fig. 5: H3K36M and H3K9M induction in HSPCs leads to widespread changes in the chromatin landscape and altered gene expression.
Fig. 6: The physiological consequences of suppressing H3K9 and H3K36 methylation are reversible both in vivo and in vitro.
Fig. 7: Prolonged expression of H3K9M/I is associated with T cell leukaemia/lymphoma.

Similar content being viewed by others

References

  1. Soshnev, A. A., Josefowicz, S. Z. & Allis, C. D. Greater than the sum of parts: complexity of the dynamic epigenome. Mol. Cell 62, 681–694 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Greer, E. L. & Shi, Y. Histone methylation: a dynamic mark in health, disease and inheritance. Nat. Rev. Genet. 13, 343–357 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Pengelly, A. R., Copur, O., Jackle, H., Herzig, A. & Muller, J. A histone mutant reproduces the phenotype caused by loss of histone-modifying factor Polycomb. Science 339, 698–699 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Maze, I., Noh, K. M., Soshnev, A. A. & Allis, C. D. Every amino acid matters: essential contributions of histone variants to mammalian development and disease. Nat. Rev. Genet. 15, 259–271 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Miller, S. A., Mohn, S. E. & Weinmann, A. S. Jmjd3 and UTX play a demethylase-independent role in chromatin remodeling to regulate T-box family member-dependent gene expression. Mol. Cell 40, 594–605 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Shpargel, K. B., Sengoku, T., Yokoyama, S. & Magnuson, T. UTX and UTY demonstrate histone demethylase-independent function in mouse embryonic development. PLoS Genet. 8, e1002964 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Kim, E. et al. Phosphorylation of EZH2 activates STAT3 signaling via STAT3 methylation and promotes tumorigenicity of glioblastoma stem-like cells. Cancer Cell 23, 839–852 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Xu, K. et al. EZH2 oncogenic activity in castration-resistant prostate cancer cells is Polycomb-independent. Science 338, 1465–1469 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lewis, P. W. et al. Inhibition of PRC2 activity by a gain-of-function H3 mutation found in pediatric glioblastoma. Science 340, 857–861 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Lu, C. et al. Histone H3K36 mutations promote sarcomagenesis through altered histone methylation landscape. Science 352, 844–849 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Behjati, S. et al. Distinct H3F3A and H3F3B driver mutations define chondroblastoma and giant cell tumor of bone. Nat. Genet. 45, 1479–1482 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Fang, D. et al. The histone H3.3K36M mutation reprograms the epigenome of chondroblastomas. Science 352, 1344–1348 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Schwartzentruber, J. et al. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature 482, 226–231 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Papillon-Cavanagh, S. et al. Impaired H3K36 methylation defines a subset of head and neck squamous cell carcinomas. Nat. Genet. 49, 180–185 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Herz, H. M. et al. Histone H3 lysine-to-methionine mutants as a paradigm to study chromatin signaling. Science 345, 1065–1070 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Jayaram, H. et al. S-adenosyl methionine is necessary for inhibition of the methyltransferase G9a by the lysine 9 to methionine mutation on histone H3. Proc. Natl Acad. Sci. USA 113, 6182–6187 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Mohammad, F. et al. EZH2 is a potential therapeutic target for H3K27M-mutant pediatric gliomas. Nat. Med. 23, 483–492 (2017).

    Article  CAS  PubMed  Google Scholar 

  18. Mohammad, F. & Helin, K. Oncohistones: drivers of pediatric cancers. Genes Dev. 31, 2313–2324 (2017).

    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. Hochedlinger, K., Yamada, Y., Beard, C. & Jaenisch, R. Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell 121, 465–477 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Wray, J. et al. Inhibition of glycogen synthase kinase-3 alleviates Tcf3 repression of the pluripotency network and increases embryonic stem cell resistance to differentiation. Nat. Cell Biol. 13, 838–845 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zhang, Y. et al. H3K36 histone methyltransferase Setd2 is required for murine embryonic stem cell differentiation toward endoderm. Cell Rep. 8, 1989–2002 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Inagawa, M. et al. Histone H3 lysine 9 methyltransferases, G9a and GLP are essential for cardiac morphogenesis. Mech. Dev. 130, 519–531 (2013).

    Article  CAS  PubMed  Google Scholar 

  24. Bilodeau, S., Kagey, M. H., Frampton, G. M., Rahl, P. B. & Young, R. A. SetDB1 contributes to repression of genes encoding developmental regulators and maintenance of ES cell state. Genes Dev. 23, 2484–2489 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Zuo, X. et al. The histone methyltransferase Setd2 is required for expression of acrosin-binding protein 1and protamines and essential for spermiogenesis in mice. J. Biol. Chem. 293, 9188–9197 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Sitnicka, E. et al. Key role of flt3 ligand in regulation of the common lymphoid progenitor but not in maintenance of the hematopoietic stem cell pool. Immunity 17, 463–472 (2002).

    Article  CAS  PubMed  Google Scholar 

  27. Adolfsson, J. et al. Identification of Flt3+ lympho-myeloid stem cells lacking erythro-megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 121, 295–306 (2005).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, Y. L. et al. Setd2 deficiency impairs hematopoietic stem cell self-renewal and causes malignant transformation. Cell Res. 28, 476–490 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hock, H. et al. Gfi-1 restricts proliferation and preserves functional integrity of haematopoietic stem cells. Nature 431, 1002–1007 (2004).

    Article  CAS  PubMed  Google Scholar 

  30. Hock, H. et al. Intrinsic requirement for zinc finger transcription factor Gfi-1 in neutrophil differentiation. Immunity 18, 109–120 (2003).

    Article  CAS  PubMed  Google Scholar 

  31. Ye, M. et al. C/EBPa controls acquisition and maintenance of adult haematopoietic stem cell quiescence. Nat. Cell Biol. 15, 385–394 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, D. E. et al. Absence of granulocyte colony-stimulating factor signaling and neutrophil development in CCAAT enhancer binding protein alpha-deficient mice. Proc. Natl Acad. Sci. USA 94, 569–574 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Ng, S. Y., Yoshida, T., Zhang, J. & Georgopoulos, K. Genome-wide lineage-specific transcriptional networks underscore Ikaros-dependent lymphoid priming in hematopoietic stem cells. Immunity 30, 493–507 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pronk, C. J. et al. Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. Cell Stem Cell 1, 428–442 (2007).

    Article  CAS  PubMed  Google Scholar 

  35. Yuan, W. et al. H3K36 methylation antagonizes PRC2-mediated H3K27 methylation. J. Biol. Chem. 286, 7983–7989 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Baubec, T. et al. Genomic profiling of DNA methyltransferases reveals a role for DNMT3B in genic methylation. Nature 520, 243–247 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Hardy, R. R., Carmack, C. E., Shinton, S. A., Kemp, J. D. & Hayakawa, K. Resolution and characterization of pro-B and pre-pro-B cell stages in normal mouse bone marrow. J. Exp. Med. 173, 1213–1225 (1991).

    Article  CAS  PubMed  Google Scholar 

  38. Lehnertz, B. et al. H3(K27M/I) mutations promote context-dependent transformation in acute myeloid leukemia with RUNX1 alterations. Blood 130, 2204–2214 (2017).

    Article  CAS  PubMed  Google Scholar 

  39. Peters, A. H. et al. Loss of the Suv39h histone methyltransferases impairs mammalian heterochromatin and genome stability. Cell 107, 323–337 (2001).

    Article  CAS  PubMed  Google Scholar 

  40. Zhuang, L. et al. Depletion of Nsd2-mediated histone H3K36 methylation impairs adipose tissue development and function. Nat. Commun. 9, 1796 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Shan, C. M. et al. A histone H3K9M mutation traps histone methyltransferase Clr4 to prevent heterochromatin spreading. eLife 5, e17903 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Yang, S. et al. Molecular basis for oncohistone H3 recognition by SETD2 methyltransferase. Genes Dev. 30, 1611–1616 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Justin, N. et al. Structural basis of oncogenic histone H3K27M inhibition of human polycomb repressive complex 2. Nat. Commun. 7, 11316 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Jiao, L. & Liu, X. Structural basis of histone H3K27 trimethylation by an active polycomb repressive complex 2. Science 350, aac4383 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Piunti, A. et al. Therapeutic targeting of polycomb and BET bromodomain proteins in diffuse intrinsic pontine gliomas. Nat. Med. 23, 493–500 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Fang, D. et al. H3.3K27M mutant proteins reprogram epigenome by sequestering the PRC2 complex to poised enhancers. eLife 7, e36696 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Funato, K., Major, T., Lewis, P. W., Allis, C. D. & Tabar, V. Use of human embryonic stem cells to model pediatric gliomas with H3.3K27M histone mutation. Science 346, 1529–1533 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Blanpain, C. & Fuchs, E. Stem cell plasticity. Plasticity of epithelial stem cells in tissue regeneration. Science 344, 1242281 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Booth, L. N. & Brunet, A. The aging epigenome. Mol. Cell 62, 728–744 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. de Graaf, C. A. et al. Haemopedia: an expression atlas of murine hematopoietic cells. Stem Cell Rep. 7, 571–582 (2016).

    Article  Google Scholar 

  51. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Nagy, A., Rossant, J., Nagy, R., Abramow-Newerly, W. & Roder, J. C. Derivation of completely cell culture-derived mice from early-passage embryonic stem cells. Proc. Natl Acad. Sci. USA 90, 8424–8428 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Eggan, K. et al. Hybrid vigor, fetal overgrowth, and viability of mice derived by nuclear cloning and tetraploid embryo complementation. Proc. Natl Acad. Sci. USA 98, 6209–6214 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  55. Anders, S., Pyl, P. T. & Huber, W. HTSeq-a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  PubMed  Google Scholar 

  56. 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 

  57. Lo, Y. H. et al. Transcriptional regulation by ATOH1 and its target SPDEF in the intestine. Cell. Mol. Gastroenterol. Hepatol. 3, 51–71 (2017).

    Article  PubMed  Google Scholar 

  58. Green, C. D. et al. A Comprehensive Roadmap of Murine Spermatogenesis Defined by Single-Cell RNA-Seq. Dev. Cell 46, 651–667.e10 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Haber, A. L. et al. A single-cell survey of the small intestinal epithelium. Nature 551, 333–339 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank M. Handley, A. Galvin, M. Gesner and E. Surette of the MGH/HSCI Rodent Histopathology Core for technical assistance. They also thank members of the Hochedlinger Lab for helpful discussions. A.J.H. is supported by an American Cancer Society—New England Division—Ellison Foundation Postdoctoral Fellowship (PF-15-130-01-DDC). B.D.S. was supported by an EMBO long-term fellowship (no. ALTF 1143-2015) and a MGH Tosteson and FMD postdoctoral fellowship. K.H. was supported by funds from the MGH, NIH (R01 HD058013-06) and the Gerald and Darlene Jordan Chair in Regenerative Medicine. H.H. was supported by a Hyundai Hope on Wheels Scholar Grant. J.B. is grateful for support from the NIH (1F32HD078029-01A1). R.I.S. was supported by funds from the NIH (P30-DK40561).

Author information

Authors and Affiliations

Authors

Contributions

J.B., H.H. and K.H. conceived the study and wrote the manuscript. J.B., B.A.S., A.J.H., J. Choi, J. Charlton, J.W.S., A.C., I.S.K., B.D.S., A.M., B.B. and R.M.W. designed and performed the experiments and analysed the data. F.J., A.A. and R.I.S. the performed bioinformatics analysis.

Corresponding authors

Correspondence to Hanno Hock or Konrad Hochedlinger.

Ethics declarations

Competing interests

The authors declare no competing interests.

Extended data

Extended Data Fig. 1 Expression of H3K9M and H3K36M suppresses methylation and blocks differentiation of pluripotent stem cells.

(a) Western blot analysis for tri-methyl H3K9 and H3K36 with and without mutant histone expression in mouse ES cells. (b) Pie charts showing the distribution of ATAC-seq peaks in EBs expressing H3, H3K9M, and H3K36M. (c) Images of teratomas derived from ES cells expressing H3, H3K9M, or H3K36M. Scale bar = 200 μm; endo = endoderm, meso = mesoderm, ecto = ectoderm. (d) qRT-PCR analysis for knockdown of H3K9 and H3K36 KMTs. One sample is shown. (e) Flow cytometry for Rex1-GFP at days 2 and 3 of embryoid body formation. (f) Western blot analysis showing H3K36me3 levels following knockdown of H3K36 KMTs. (g) Western blot analysis showing H3K9me3 levels following knockdown of H3K9 KMTs. See source data for full membrane Western blot images. Data in a,c,e,f,g are representative of 3 independent experiments.

Source data

Source data

Extended Data Fig. 2 In vivo expression of H3K9M and H3K36M disrupts tissue homeostasis.

(a) Histological analysis of testis in mice induced for 4 weeks. Scale bar = 20 μm. (b) GSEA based on RNA-seq data from testes isolated from H3K9M and H3K36M mice induced for 4 weeks (H3, n = 2; H3K9M, n = 2; H3K36M, n = 2). Enrichment is shown for transcriptional signatures58 related to spermatogenesis. Statistics were generated in accordance with the published GSEA algorithm51 (c) Gene tracks showing expression for genes characteristic of spermatogonia (left panel) or spermatocytes (right panel). (d) Histological analysis of intestine in mice induced for 4 weeks. Black and white arrowheads indicate goblet cells and paneth cells, respectively. Scale bar = 20 μm. (e) Staining for markers of Paneth cells (Lysozyme; left panel) and goblet cells (periodic acid Schiff; right panel) (f) GSEA based on RNA-seq data from intestine isolated from H3K9M and H3K36M mice induced for 4 weeks (H3, n = 2; H3K9M, n = 2; H3K36M, n = 2). Enrichment is shown for transcriptional signatures related to secretory lineages57 and Goblet cells59. Statistics were generated in accordance with the published GSEA algorithm51. (g) Gene tracks showing expression for genes characteristic of secretory lineages (left panel) or goblet cells (right panel). (h) Tibia from mice expressing mutant histones for 4 weeks. Scale bar = 5 mm. Data in a,d,e,h are representative of 3 independent experiments.

Extended Data Fig. 3 H3K9M and H3K36M induction cause abnormalities in the erythroid, megakaryocyte, granulocyte and T cell lineages.

(a) cKit expression in early erythroid progenitors. Data are representative of three independent experiments. (b) Quantification of cKit positive erythroid progenitors (right panel). Columns represent the mean and error bars represent standard deviation of the mean for biological replicates (rtTA, n = 6; H3K9M, n = 3; H3K36M, n = 3). Statistical significance was determined using a two-tailed unpaired Student’s t-test. (c) Time course analysis for hemoglobin assessed by CBC. Columns represent the mean and error bars represent standard deviation of the mean for biological replicates (rtTA, n = 5; H3K9M, n = 4; H3K36M, n = 3). Statistical significance was determined using the Holm-Sidak method. (d) An infarcted spleen from a H3K9M mouse induced for 4 weeks. Scale bar = 5 mm. Infarctions were present in 2 out of 3 mice inspected. (e) Flow cytometry analysis for granulocyte cells. Gr1+ cells are shown in green. (f) Granularity of transgenic cells assessed by side scatter. (g) Cytospin images of Gr1+ granulocytes sorted from H3K36M and rtTA mice. Scale bar = 5 μm. (h) Flow cytometry analysis for thymocytes. Frequencies for each cell type are indicated as a percentage of the parent gate. (i) Quantification of CD4/CD8 double negative thymocytes (left panel) and double positive CD4/CD8 thymocytes (right panel). Columns represent the mean and error bars represent standard deviation of the mean for biological replicates (rtTA, n = 6; H3K9M, n = 3; H3K36M, n = 3). Statistical significance was determined using a two-tailed unpaired Student’s t-test. Data in e,f,g,h are representative of 3 independent experiments.

Source data

Extended Data Fig. 4 Mutant histone expression impacts HSC function in competitive bone marrow transplantation assays.

(a) Frequency of granulocytes derived from donor bone marrow (CD45.2+) harboring control or mutant histones before and after 12 weeks of induction with doxycyline. Columns represent the mean and error bars represent standard deviation of the mean for biological replicates (n = 4). Statistical significance was determined using a two-tailed unpaired Student’s t-test. (b) Flow cytometry analysis for donor-derived, CD45.2+ LKS cells, hematopoietic progenitor cells, and stem cells at the conclusion of competitive bone marrow transplant assays. Frequencies for each cell type are indicated as a percentage of the parent gate. (c) Flow cytometry showing granularity of both wild-type competitor marrow (CD45.1) and marrow expressing histone transgenes (CD45.2) as assessed by side scatter. (d) Brightfield images of colonies in methylcellulose. Black arrowheads indicate megakaryocytes. Scale bar = 200 μm or 50 μm. (e) May Grünwald Giemsa stains of cells grown in methylcellulose. Representative images are shown for rtTA, H3K9M, and H3K36M. Scale bar = 20 μm. Black arrowheads indicate megakaryocytes, white arrowheads indicate granulocytes, and red arrowheads indicate erythroid cells. Data in b,c,d,e are representative of 3 independent experiments.

Source data

Extended Data Fig. 5 Mutant histone expression impacts HSC gene expression.

(a) GSEA analysis based on RNA-seq data for hematopoietic stem and progenitor cells sorted from H3K9M and H3K36M mice induced for 4 weeks (H3, n = 2; H3K9M, n = 2; H3K36M, n = 2). Enrichment is shown for transcriptional signatures related to Lymphoid/Myeloid progenitors (CMP, LMPP, and proB)33 and B cells50 (K9M) as well as erythroid cells33 and B cells50 (K36M). Statistics were generated in accordance with the published GSEA algorithm51. (b) Principal component analysis based on the union of differentially expressed genes between rtTA control and H3K9M or H3K36M samples. Each circle represents a biological replicate (rtTA, n = 3; H3K9M, n = 3; H3K36M, n = 3) while squares represent the mean. (c) Expression (RPKM) values for select genes relative to average expression in rtTA replicates (shown in log2 scale) for biological triplicates from LKS cells sorted at day 7 of induction.

Extended Data Fig. 6 H3K36M/H3K9M induction change the chromatin landscape in HSPCs.

(a) Distribution of H3K9me3 across genomic elements. (b) Heat-maps showing H3K9me3 methylation levels genome-wide following four weeks of H3 or H3K9M induction (averages, two replicates). (c) Representative tracks for loci with decreased H3K9me3 signal following H3K9M expression. (d) RNA-seq scatter plot based on RPKM. Red lines marks 1.5-fold difference between WT and K9M, gray line marks RPKM = 1 (cutoff for expressed repeats). Differentially expressed repeats (1.5-fold, FDR < 0.1; RNA-Seq data) are marked in orange and green dots. Red circles mark loci where H3K9me3 is differentially bound. Repeats overlapping protein-coding exons were excluded. (e) Heat-maps showing H3K36me3 methylation levels genome-wide following four weeks of H3 or H3K36M induction (averages, two replicates). (f) Distribution of H3K36me3 across genomic elements. (g) Levels of H3K36me3 in genes with increased expression, decreased expression, and all genes in H3K36M vs control HSPCs. The center bar represents the mean and the whiskers represent the standard deviation of the mean. (h) Replicate heat-maps showing H3K27me3 methylation for genes that are downregulated following H3K36M induction. (i) Correlation plot comparing H3K27me3 levels in H3K36M vs. H3 HSPCs. Levels of H3K36me3 overlaid on data points. (j) Meta-analysis showing H3K27me3 levels for a 200 kb region surrounding genes that are downregulated with H3K36M expression (top) and at random regions (bottom). (k) ATAC-seq scatter plot. Loci with reduced H3K36me3 following H3K36M expression are in blue. (l) Replicate tracks for Gfi1 in Fig. 5g. (m) Representative tracks for Prdm5, which is downregulated following H3K36M expression. (n) Correlation plots comparing gene body methylation between H3 or H3K36M HSPCs. Significant differences in green (left), hematopoietic genes in blue (right). (o) Gene tracks for Gfi1 showing DNA methylation in H3 or H3K36M HSPCs. (p) Replicate heat-maps showing H3K36me3 methylation at gene bodies for genes upregulated in H3K36M HSPCs. (q) Replicate tracks for Klf5 in Fig. 5h. (r) Representative tracks for Zfpm1, which is upregulated following H3K36M expression. N numbers in d represent repetitive elements. N numbers in g,n represent genes. All n numbers are stated in the figure.

Extended Data Fig. 7 Reversibility of K-to-M dependent phenotypes.

(a) Histological analysis of testis in mice induced for 4 weeks with an additional 4 weeks of dox withdrawal. Scale bar = 50 μm. (b) Histological analysis of intestine in mice induced for 4 weeks with an additional 4 weeks of dox withdrawal. Black and white arrowheads indicate goblet cells and paneth cells, respectively. Scale bar = 50 μm. (c) Brightfield images of colonies in methylcellulose with dox removal. Black arrowheads indicate megakaryocytes. Scale bar = 200 μm or 50 μm. (d) Western blot analysis for tri-methyl H3K9 and H3K36 from EBs induced for 3 days with dox and EBs induced for 3 days with dox followed by 3 days of dox withdrawal. (e) Western blot analysis for H3K9me3 and H3K36me3 from HSPCs in mice induced for 7 days with dox and mice induced for 7 days with dox followed by 7 days of dox withdrawal. See source data for full membrane Western blot images. Data in a-e are representative of 3 independent experiments.

Source data

Extended Data Fig. 8 K9I mice phenocopy K9M mice.

(a) Flow cytometry analysis of megakaryocyte progenitors (MkPs) cells, 4 weeks of dox. Frequencies as percentages of the parent gate. (b) Quantification of MkPs in bone marrow. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 3, biological replicates). (c) Platelet counts after 4 weeks of dox. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 3, biological replicates). (d) Flow cytometry of B cells in the bone marrow, 4 weeks of dox. Frequencies as percentages of the parent gates. (e) Quantification of mature B cells and IGM+ B cells. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 2, biological replicates). (f) Flow cytometry analysis of B cell progenitors in bone marrow, 4 weeks of dox. Frequencies as percentages of parent gates. (g) Quantification of ProB cells and PreB cells. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 2, biological replicates). (h) Flow cytometry analysis of LKS cells. Frequencies as percentages of parent gates. (i) Quantification of LKS cells in bone marrow. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 3, biological replicates). (j) Flow cytometry of stem cell populations gated on LKS cells. Frequencies indicated as percentages of parent gates. (k) Quantification of LMPPs, defined as Flt3high cells (top 25% of Flt3+ cells set on rtTA control). Columns represent means and error bars standard deviation (rtTA, n = 1; H3K9I, n = 3, biological replicates). (l) Flow cytometry of SLAM markers gated on LKS cells. Frequencies as percentages of parent gates. (m) Quantification of MPPs (left panel) and long-term HSCs (right panel) in bone marrow. Columns represent the mean +/− standard deviation (rtTA, n = 1; H3K9I, n = 3 biological replicates). Data in a,d,f,h,j,l are representative of 3 independent experiments.

Source data

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

Brumbaugh, J., Kim, I.S., Ji, F. et al. Inducible histone K-to-M mutations are dynamic tools to probe the physiological role of site-specific histone methylation in vitro and in vivo. Nat Cell Biol 21, 1449–1461 (2019). https://doi.org/10.1038/s41556-019-0403-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-019-0403-5

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