A germline JAK2 SNP is associated with predisposition to the development of JAK2V617F-positive myeloproliferative neoplasms (original) (raw)

Nature Genetics volume 41, pages 455–459 (2009)Cite this article

Abstract

Polycythemia vera, essential thrombocythemia and primary myelofibrosis are myeloproliferative neoplasms (MPN) characterized by multilineage clonal hematopoiesis1,2,3,4,5. Given that the identical somatic activating mutation in the JAK2 tyrosine kinase gene (_JAK2_V617F) is observed in most individuals with polycythemia vera, essential thrombocythemia and primary myelofibrosis6,7,8,9,10, there likely are additional genetic events that contribute to the pathogenesis of these phenotypically distinct disorders. Moreover, family members of individuals with MPN are at higher risk for the development of MPN, consistent with the existence of MPN predisposition loci11. We hypothesized that germline variation contributes to MPN predisposition and phenotypic pleiotropy. Genome-wide analysis identified an allele in the JAK2 locus (rs10974944) that predisposes to the development of _JAK2_V617F-positive MPN, as well as three previously unknown MPN modifier loci. We found that _JAK2_V617F is preferentially acquired in cis with the predisposition allele. These data suggest that germline variation is an important contributor to MPN phenotype and predisposition.

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

Access options

Subscribe to this journal

Receive 12 print issues and online access

$209.00 per year

only $17.42 per issue

Buy this article

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

Additional access options:

Similar content being viewed by others

References

  1. Adamson, J.W., Fialkow, P.J., Murphy, S., Prchal, J.F. & Steinmann, L. Polycythemia vera: stem-cell and probable clonal origin of the disease. N. Engl. J. Med. 295, 913–916 (1976).
    Article CAS Google Scholar
  2. Gilliland, D.G., Blanchard, K.L., Levy, J., Perrin, S. & Bunn, H.F. Clonality in myeloproliferative disorders: analysis by means of the polymerase chain reaction. Proc. Natl. Acad. Sci. USA 88, 6848–6852 (1991).
    Article CAS Google Scholar
  3. El Kassar, N., Hetet, G., Li, Y., Briere, J. & Grandchamp, B. Clonal analysis of haemopoietic cells in essential thrombocythaemia. Br. J. Haem. 90, 131–137 (1995).
    Article CAS Google Scholar
  4. Tsukamoto, N. et al. Clonality in chronic myeloproliferative disorders defined by X-chromosome linked probes: demonstration of heterogeneity in lineage involvement. Br. J. Haematol. 86, 253–258 (1994).
    Article CAS Google Scholar
  5. Jamieson, C.H. et al. The JAK2 V617F mutation occurs in hematopoietic stem cells in polycythemia vera and predisposes toward erythroid differentiation. Proc. Natl. Acad. Sci. USA 103, 6224–6229 (2006).
    Article CAS Google Scholar
  6. James, C. et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 434, 1144–1148 (2005).
    Article CAS Google Scholar
  7. Baxter, E.J. et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365, 1054–1061 (2005).
    Article CAS Google Scholar
  8. Kralovics, R. et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N. Engl. J. Med. 352, 1779–1790 (2005).
    Article CAS Google Scholar
  9. Zhao, R. et al. Identification of an acquired JAK2 mutation in polycythemia vera. J. Biol. Chem. 280, 22788–22792 (2005).
    Article CAS Google Scholar
  10. Levine, R.L. et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7, 387–397 (2005).
    Article CAS Google Scholar
  11. Landgren, O. et al. Increased risks of polycythemia vera, essential thrombocythemia, and myelofibrosis among 24577 first-degree relatives of 11039 patients with myeloproliferative neoplasms in Sweden. Blood 112, 2199–2204 (2008).
    Article CAS Google Scholar
  12. Pardanani, A., Fridley, B.L., Lasho, T.L., Gilliland, D.G. & Tefferi, A. Host genetic variation contributes to phenotypic diversity in myeloproliferative disorders. Blood 111, 2785–2789 (2008).
    Article CAS Google Scholar
  13. Cario, H., Goerttler, P.S., Steimle, C., Levine, R.L. & Pahl, H.L. The JAK2V617F mutation is acquired secondary to the predisposing alteration in familial polycythaemia vera. Br. J. Haematol. 130, 800–801 (2005).
    Article CAS Google Scholar
  14. Bellanne-Chantelot, C. et al. Genetic and clinical implications of the Val617Phe JAK2 mutation in 72 families with myeloproliferative disorders. Blood 108, 346–352 (2006).
    Article CAS Google Scholar
  15. Pietra, D. et al. Somatic mutations of JAK2 exon 12 in patients with JAK2 (V617F)-negative myeloproliferative disorders. Blood 111, 1686–1689 (2008).
    Article CAS Google Scholar
  16. Scott, L.M., Scott, M.A., Campbell, P.J. & Green, A.R. Progenitors homozygous for the V617F mutation occur in most patients with polycythemia vera, but not essential thrombocythemia. Blood 108, 2435–2437 (2006).
    Article CAS Google Scholar
  17. Scott, L.M. et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N. Engl. J. Med. 356, 459–468 (2007).
    Article CAS Google Scholar
  18. Di Bernardo, M.C. et al. A genome-wide association study identifies six susceptibility loci for chronic lymphocytic leukemia. Nat. Genet. 40, 1204–1210 (2008).
    Article CAS Google Scholar
  19. Hemminki, K., Forsti, A. & Bermejo, J.L. The 'common disease-common variant' hypothesis and familial risks. PLoS ONE 3, e2504 (2008).
    Article Google Scholar
  20. Hemminki, K., Forsti, A. & Lorenzo Bermejo, J. New cancer susceptibility loci: population and familial risks. Int. J. Cancer 123, 1726–1729 (2008).
    Article CAS Google Scholar
  21. Laken, S.J. et al. Familial colorectal cancer in Ashkenazim due to a hypermutable tract in APC. Nat. Genet. 17, 79–83 (1997).
    Article CAS Google Scholar
  22. Bercovich, D. et al. Mutations of JAK2 in acute lymphoblastic leukaemias associated with Down's syndrome. Lancet 372, 1484–1492 (2008).
    Article CAS Google Scholar
  23. Mercher, T. et al. JAK2T875N is a novel activating mutation that results in myeloproliferative disease with features of megakaryoblastic leukemia in a murine bone marrow transplantation model. Blood 108, 2770–2779 (2006).
    Article CAS Google Scholar
  24. The Cancer Genome Atlas Research Network. Comprehensive genomic characterization defines human glioblastoma genes and core pathways. Nature 455, 1061–1068 (2008).
  25. Levine, R.L. et al. X-inactivation-based clonality analysis and quantitative JAK2V617F assessment reveal a strong association between clonality and JAK2V617F in PV but not ET/MMM, and identifies a subset of JAK2V617F-negative ET and MMM patients with clonal hematopoiesis. Blood 107, 4139–4141 (2006).
    Article CAS Google Scholar
  26. Purcell, S. et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am. J. Hum. Genet. 81, 559–575 (2007).
    Article CAS Google Scholar
  27. The Wellcome Trust Case Control Consortium. Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls. Nature 447, 661–678 (2007).
  28. Price, A.L. et al. Principal components analysis corrects for stratification in genome-wide association studies. Nat. Genet. 38, 904–909 (2006).
    Article CAS Google Scholar

Download references

Acknowledgements

We would like to acknowledge the subjects who have contributed to our understanding of these disorders. We thank S. Thomas, I. Dolgalev and T. Landers for assistance with high-throughput resequencing, A. Viale for assistance with JAK2 expression analysis, and T. Kirchhoff for advice and suggestions. This study makes use of data generated by the Wellcome Trust Case-Control Consortium; a full list of the investigators who contributed to the generation of the data are available from http://www.wtccc.org.uk and funding was provided by the Wellcome Trust under award 076113. This work was supported by grants from the National Institutes of Health, the Starr Cancer Consortium, the Myeloproliferative Disorders Foundation, the Howard Hughes Medical Institute, the Doris Duke Charitable Foundation and the Kristen Amico Sesselman Leukemia Research Fund. O.K. is supported by a grant from the Academy of Finland. D.G.G. is an Investigator of the Howard Hughes Medical Institute and is a Doris Duke Charitable Foundation Distinguished Clinical Scientist. Work in the laboratory of R.J.K. is supported by Memorial Sloan Kettering Cancer Center through US National Institutes of Health grant P30 CA008748. R.L.L. is an Early Career Award recipient of the Howard Hughes Medical Institute and a Clinical Scientist Development Award recipient of the Doris Duke Charitable Foundation and is the Geoffrey Beene Junior Chair at Memorial Sloan Kettering Cancer Center.

Author information

Author notes

  1. Outi Kilpivaara and Semanti Mukherjee: These authors contributed equally to this work.

Authors and Affiliations

  1. Human Oncology and Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
    Outi Kilpivaara, Alison M Schram, Sachie Marubayashi, Adriana Heguy & Ross L Levine
  2. Cancer Biology and Genetics Program, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
    Semanti Mukherjee & Robert J Klein
  3. Gerstner Sloan-Kettering Graduate School of Biomedical Sciences, New York, New York, USA
    Semanti Mukherjee
  4. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
    Martha Wadleigh, Ann Mullally, Adam Bass, Richard M Stone & D Gary Gilliland
  5. Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
    Ann Mullally, Benjamin L Ebert & D Gary Gilliland
  6. Broad Institute of Harvard and MIT, Cambridge, Massachusetts, USA
    Benjamin L Ebert, Adam Bass & D Gary Gilliland
  7. Department of Leukemia, M.D. Anderson Cancer Center, Houston, Texas, USA
    Guillermo Garcia-Manero & Hagop Kantarjian
  8. Department of Medicine, Clinical Genetics Service, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
    Kenneth Offit
  9. Howard Hughes Medical Institute, Harvard Medical School, Boston, Massachusetts, USA
    D Gary Gilliland
  10. Harvard Stem Cell Institute, Boston, Massachusetts, USA
    D Gary Gilliland
  11. Department of Medicine, Leukemia Service, Memorial Sloan-Kettering Cancer Center, New York, New York, USA
    Ross L Levine

Authors

  1. Outi Kilpivaara
    You can also search for this author inPubMed Google Scholar
  2. Semanti Mukherjee
    You can also search for this author inPubMed Google Scholar
  3. Alison M Schram
    You can also search for this author inPubMed Google Scholar
  4. Martha Wadleigh
    You can also search for this author inPubMed Google Scholar
  5. Ann Mullally
    You can also search for this author inPubMed Google Scholar
  6. Benjamin L Ebert
    You can also search for this author inPubMed Google Scholar
  7. Adam Bass
    You can also search for this author inPubMed Google Scholar
  8. Sachie Marubayashi
    You can also search for this author inPubMed Google Scholar
  9. Adriana Heguy
    You can also search for this author inPubMed Google Scholar
  10. Guillermo Garcia-Manero
    You can also search for this author inPubMed Google Scholar
  11. Hagop Kantarjian
    You can also search for this author inPubMed Google Scholar
  12. Kenneth Offit
    You can also search for this author inPubMed Google Scholar
  13. Richard M Stone
    You can also search for this author inPubMed Google Scholar
  14. D Gary Gilliland
    You can also search for this author inPubMed Google Scholar
  15. Robert J Klein
    You can also search for this author inPubMed Google Scholar
  16. Ross L Levine
    You can also search for this author inPubMed Google Scholar

Contributions

The study was designed by O.K., S. Mukherjee, R.J.K. and R.L.L. with advice from K.O. SNP arrays were performed and analyzed by A.B., B.L.E. and R.L.L, and analysis of SNP array data for modifier and predisposition loci was performed by S. Mukherjee and R.J.K. Genotyping, sequence analysis and real-time PCR assays were performed by O.K., A.M.S., S. Marubayashi, A.H. and R.L.L. Principal component analysis was done by S. Mukherjee and R.J.K. Identification of subjects, sample collection and phenotypic assessment were done by M.W., A.M., G.G.-M., H.K., R.M.S, D.G.G. and R.L.L. The paper was written by O.K., S. Mukherjee, K.O., D.G.G., R.J.K. and R.L.L. All authors discussed the results and commented on the manuscript.

Corresponding authors

Correspondence toRobert J Klein or Ross L Levine.

Supplementary information

Rights and permissions

About this article

Cite this article

Kilpivaara, O., Mukherjee, S., Schram, A. et al. A germline JAK2 SNP is associated with predisposition to the development of _JAK2_V617F-positive myeloproliferative neoplasms.Nat Genet 41, 455–459 (2009). https://doi.org/10.1038/ng.342

Download citation

This article is cited by

Associated content