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.

  • Letter
  • Published:

IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics

This article has been updated

Abstract

Mutations in the IDH1 and IDH2 genes encoding isocitrate dehydrogenases are frequently found in human glioblastomas1 and cytogenetically normal acute myeloid leukaemias (AML)2. These alterations are gain-of-function mutations in that they drive the synthesis of the ‘oncometabolite’ R-2-hydroxyglutarate (2HG)3. It remains unclear how IDH1 and IDH2 mutations modify myeloid cell development and promote leukaemogenesis. Here we report the characterization of conditional knock-in (KI) mice in which the most common IDH1 mutation, IDH1(R132H), is inserted into the endogenous murine Idh1 locus and is expressed in all haematopoietic cells (Vav-KI mice) or specifically in cells of the myeloid lineage (LysM-KI mice). These mutants show increased numbers of early haematopoietic progenitors and develop splenomegaly and anaemia with extramedullary haematopoiesis, suggesting a dysfunctional bone marrow niche. Furthermore, LysM-KI cells have hypermethylated histones and changes to DNA methylation similar to those observed in human IDH1- or IDH2-mutant AML. To our knowledge, our study is the first to describe the generation and characterization of conditional IDH1(R132H)-KI mice, and also the first report to demonstrate the induction of a leukaemic DNA methylation signature in a mouse model. Our report thus sheds light on the mechanistic links between IDH1 mutation and human AML.

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: LysM-KI mice show age-dependent splenomegaly and decreased bone marrow cellularity.
Figure 2: LysM-KI mice show age-dependent increases in lineage-restricted progenitors and extramedullary haematopoiesis.
Figure 3: Altered methylation of DNA and histones in LysM-KI cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

Data deposits

The microarray and sequencing data have been deposited in the Gene ExpressionOmnibus of the National Center for Biotechnical Information with the accession numbers GSE38589 and GSE38687.

Change history

  • 29 August 2012

    The spelling of an author name (S.M.S.) was corrected

References

  1. Parsons, D. W. et al. An integrated genomic analysis of human glioblastoma multiforme. Science 321, 1807–1812 (2008)

    Article  ADS  CAS  Google Scholar 

  2. Mardis, E. R. et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. N. Engl. J. Med. 361, 1058–1066 (2009)

    Article  CAS  Google Scholar 

  3. Dang, L. et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature 462, 739–744 (2009)

    Article  ADS  CAS  Google Scholar 

  4. Xu, W. et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of α-ketoglutarate-dependent dioxygenases. Cancer Cell 19, 17–30 (2011)

    Article  CAS  Google Scholar 

  5. Figueroa, M. E. et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 18, 553–567 (2010)

    Article  CAS  Google Scholar 

  6. Chowdhury, R. et al. The oncometabolite 2-hydroxyglutarate inhibits histone lysine demethylases. EMBO Rep. 12, 463–469 (2011)

    Article  CAS  Google Scholar 

  7. Clausen, B. E. et al. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res. 8, 265–277 (1999)

    Article  CAS  Google Scholar 

  8. Ye, M. et al. Hematopoietic stem cells expressing the myeloid lysozyme gene retain long-term, multilineage repopulation potential. Immunity 19, 689–699 (2003)

    Article  CAS  Google Scholar 

  9. Goardon, N. et al. Coexistence of LMPP-like and GMP-like leukemia stem cells in acute myeloid leukemia. Cancer Cell 19, 138–152 (2011)

    Article  CAS  Google Scholar 

  10. Holmes R & Zúñiga-Pflücker J.-C The OP9–DL1 system: generation of T-lymphocytes from embryonic or hematopoietic stem cells in vitro. Cold Spring Harb. Protoc. http://dx.doi.org/10.1101/pdb.prot5156 (2009)

  11. de Boer, J. et al. Transgenic mice with hematopoietic and lymphoid specific expression of Cre. Eur. J. Immunol. 33, 314–325 (2003)

    Article  CAS  Google Scholar 

  12. Ito, K. et al. Reactive oxygen species act through p38 MAPK to limit the lifespan of hematopoietic stem cells. Nature Med. 12, 446–451 (2006)

    Article  CAS  Google Scholar 

  13. Tothova, Z. et al. FoxOs are critical mediators of hematopoietic stem cell resistance to physiologic oxidative stress. Cell 128, 325–339 (2007)

    Article  CAS  Google Scholar 

  14. Callens, C. et al. Targeting iron homeostasis induces cellular differentiation and synergizes with differentiating agents in acute myeloid leukemia. J. Exp. Med. 207, 731–750 (2010)

    Article  CAS  Google Scholar 

  15. Eliasson, P. & Jönsson, J. I. The hematopoietic stem cell niche: low in oxygen but a nice place to be. J. Cell. Physiol. 222, 17–22 (2010)

    Article  CAS  Google Scholar 

  16. Zhao, S. et al. Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1α. Science 324, 261–265 (2009)

    Article  ADS  CAS  Google Scholar 

  17. Noushmehr, H. et al. Identification of a CpG island methylator phenotype that defines a distinct subgroup of glioma. Cancer Cell 17, 510–522 (2010)

    Article  CAS  Google Scholar 

  18. Akalin, A. et al. Base-pair resolution DNA methylation sequencing reveals profoundly divergent epigenetic landscapes in acute myeloid leukemia. PLoS Genet. (in the press)

  19. Campbell, C. et al. Signal control of hematopoietic stem cell fate: Wnt, Notch, and Hedgehog as the usual suspects. Curr. Opin. Hematol. 15, 319–325 (2008)

    Article  CAS  Google Scholar 

  20. Heidel, F. H. et al. Self-renewal related signaling in myeloid leukemia stem cells. Int. J. Hematol. 94, 109–117 (2011)

    Article  Google Scholar 

  21. Klinakis, A. et al. A novel tumour-suppressor function for the Notch pathway in myeloid leukaemia. Nature 473, 230–233 (2011)

    Article  ADS  CAS  Google Scholar 

  22. Söderberg, S. S. et al. Complex and context dependent regulation of hematopoiesis by TGF-β superfamily signaling. Ann. NY Acad. Sci. 1176, 55–69 (2009)

    Article  ADS  Google Scholar 

  23. Patel, K. P. et al. Acute myeloid leukemia with IDH1 or IDH2 mutation: frequency and clinicopathologic features. Am. J. Clin. Pathol. 135, 35–45 (2011)

    Article  CAS  Google Scholar 

  24. Patnaik, M. M. et al. Differential prognostic effect of IDH1 versus IDH2 mutations in myelodysplastic syndromes: a Mayo Clinic Study of 277 patients. Leukemia 26, 101–105 (2012)

    Article  CAS  Google Scholar 

  25. Munger, J. et al. Systems-level metabolic flux profiling identifies fatty acid synthesis as a target for antiviral therapy. Nature Biotechnol. 26, 1179–1186 (2008)

    Article  CAS  Google Scholar 

  26. Bajad, S. U. et al. Separation and quantitation of water soluble cellular metabolites by hydrophilic interaction chromatography-tandem mass spectrometry. J. Chromatogr. A 1125, 76–88 (2006)

    Article  CAS  Google Scholar 

  27. Meissner, A. et al. Reduced representation bisulfite sequencing for comparative high-resolution DNA methylation analysis. Nucleic Acids Res. 33, 5868–5877 (2005)

    Article  CAS  Google Scholar 

  28. Krueger, F. & Andrews, S. R. Bismark: a flexible aligner and methylation caller for Bisulfite-Seq applications. Bioinformatics 27, 1571–1572 (2011)

    Article  CAS  Google Scholar 

  29. Fujita, P. A. et al. The UCSC Genome Browser database: update 2011. Nucleic Acids Res. 39, D876–D882 (2011)

    Article  CAS  Google Scholar 

  30. Storey, J. D. & Tibshirani, R. Statistical significance for genomewide studies. Proc. Natl Acad. Sci. USA 100, 9440–9445 (2003)

    Article  ADS  MathSciNet  CAS  Google Scholar 

  31. Du, P. et al. lumi: a pipeline for processing Illumina microarray. Bioinformatics 24, 1547–1548 (2008)

    Article  CAS  Google Scholar 

  32. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc., B 57, 289–300 (1995)

    MathSciNet  MATH  Google Scholar 

  33. Smyth G. K Linear models and empirical Bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, 3 (2004)

    Article  ADS  MathSciNet  Google Scholar 

  34. Benjamini, Y. & Yekutieli, D. The control of the false discovery rate in multiple testing under dependency. Ann. Stat. 29, 1165–1188 (2001)

    Article  MathSciNet  Google Scholar 

Download references

Acknowledgements

We thank the Animal Research Colony (ARC) at the Ontario Cancer Institute for mouse care; I. Ng, A. Shahinian, J. Sylvester and S. McCracken for administrative and organizational expertise; M. Bailey and J. Tsao for technical assistance; F. Tong and R. Nayyar for assistance with flow cytometric analysis and sorting; the Weill Cornell Medical College (WCMC) Epigenomics Core Facility for technical help and expertise; G. Melino, D. Green, M. Minden, H. Chang and P. Lang for helpful discussions; J. Thomsen for figure layout and M. Saunders for scientific editing. C.B.K. and D.B. were supported in part by a Feodor-Lynen Postdoctoral Research Fellowship from the Alexander-von-Humboldt-Foundation, Germany. D.B. and A.B. were supported in part by a Fellowship from the German Research Foundation (DFG). J.C.M. is supported by a National Institute of Health grant (NIH R01AI081773) and is a Damon Runyon-Rachleff Innovation Awardee supported by the Damon Runyon Cancer Research Foundation (DRR-09-10). P.S.O. holds a Canada Research Chair in Autoimmunity and Tumor Immunity. M.E.F. is supported by the Leukemia & Lymphoma Society Special Fellow Award and a Doris Duke Clinical Scientist Development Award. A.M. is supported by an LLS SCOR grant (7132-08), a Burroughs Wellcome Clinical Translational Scientist Award and a Starr Cancer Consortium grant (I4-A442). J.-C.Z.-P. is supported by a Canada Research Chair in Developmental Immunology. This work was supported by grants from the Canadian Institutes of Health Research (CIHR) and the Ontario Ministry of Health and Long Term Care to T.W.M., and a program grant from the Terry Fox Foundation to P.S.O., J.-C.Z.-P. and T.W.M. Please note that the views expressed do not necessarily reflect those of the OMOHLTC.

Author information

Authors and Affiliations

Authors

Contributions

C.B.K., M.S. and T.W.M. initiated the project. M.S. designed the conditional IDH1(R132H)-KI mouse and generated it, as well as the IDH1(R132H)-KI embryonic stem cell clones with the help of S.S., S.M.S., A.W., J.H. and A.Y.-T.; C.B.K. designed and organized experiments involving the hematopoietic system in consultation with T.W.M., M.E.F., A.M., J.-C.Z.-P., G.R., P.S.O., C.V. and D.L.B.; C.B.K. performed the flow cytometry analyses, bone marrow transfers, cell sorting, RNA extraction and real-time PCR analyses, immunoblotting, colony formation assays and serial plating experiments. J.C.M. performed the LC-MS analyses. E.F.L., D.B., A.B. and I.S.H. helped with flow cytometric analyses and BM transfers. R.H. performed the OP9/OP9-DL1 embryonic stem cell in vitro differentiation experiments. W.Y.L. helped with flow cytometric analyses and cell culture experiments. M.E.F. and A.M. performed the DNA methylation experiments and analysed the data. C.V. performed the mRNA expression microarray experiments and analysed data. C.B.K. and T.W.M. wrote the manuscript with the help of A.B. and D.B. All authors discussed the results extensively and agree with the conclusions presented in the manuscript.

Corresponding author

Correspondence to Tak W. Mak.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-11, Supplementary Tables 1-2 and 6-7 and legends for Supplementary Tables 3, 4 and 5 (see separate files for these tables). (PDF 1609 kb)

Supplementary Table 3

This table contains differentially expressed mRNAs in LysM-KI and control LSK cells - see Supplementary Information file for full legend. (XLS 77 kb)

Supplementary Table 4

This table contains Gene Ontology categories of the differentially expressed mRNAs - see Supplementary Information file for full legend. (XLS 836 kb)

Supplementary Table 5

This table contains differentially methylated genomic regions - see Supplementary Information file for full legend. (XLS 219 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sasaki, M., Knobbe, C., Munger, J. et al. IDH1(R132H) mutation increases murine haematopoietic progenitors and alters epigenetics. Nature 488, 656–659 (2012). https://doi.org/10.1038/nature11323

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11323

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