Distinct effects of concomitant Jak2V617F expression and Tet2 loss in mice promote disease progression in myeloproliferative neoplasms.

Signaling mutations (eg, JAK2V617F) and mutations in genes involved in epigenetic regulation (eg, TET2) are the most common cooccurring classes of mutations in myeloproliferative neoplasms (MPNs). Clinical correlative studies have demonstrated that TET2 mutations are enriched in more advanced phases of MPNs such as myelofibrosis and leukemic transformation, suggesting that they may cooperate with JAK2V617F to promote disease progression. To dissect the effects of concomitant Jak2V617F expression and Tet2 loss within distinct hematopoietic compartments in vivo, we generated Jak2V617F/Tet2 compound mutant genetic mice. We found that the combination of Jak2V617F expression and Tet2 loss resulted in a more florid MPN phenotype than that seen with either allele alone. Concordant with this, we found that Tet2 deletion conferred a strong functional competitive advantage to Jak2V617F-mutant hematopoietic stem cells (HSCs). Transcriptional profiling revealed that both Jak2V617F expression and Tet2 loss were associated with distinct and nonoverlapping gene expression signatures within the HSC compartment. In aggregate, our findings indicate that Tet2 loss drives clonal dominance in HSCs, and Jak2V617F expression causes expansion of downstream precursor cell populations, resulting in disease progression through combinatorial effects. This work provides insight into the functional consequences of JAK2V617F-TET2 comutation in MPNs, particularly as it pertains to HSCs.

[1]  T. Golub,et al.  Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. , 2014, Blood.

[2]  Christian Beisel,et al.  Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. , 2014, Blood.

[3]  G. Superti-Furga,et al.  Somatic mutations of calreticulin in myeloproliferative neoplasms. , 2013, The New England journal of medicine.

[4]  J. D. Fitzpatrick,et al.  Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. , 2013, The New England journal of medicine.

[5]  M. Cazzola,et al.  Mutations and prognosis in primary myelofibrosis , 2013, Leukemia.

[6]  Benjamin J. Raphael,et al.  Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. , 2013, The New England journal of medicine.

[7]  David A. Williams,et al.  Depletion of Jak2V617F myeloproliferative neoplasm-propagating stem cells by interferon-α in a murine model of polycythemia vera. , 2013, Blood.

[8]  O. Bernard,et al.  In aggressive forms of mastocytosis, TET2 loss cooperates with c-KITD816V to transform mast cells. , 2012, Blood.

[9]  B. Ebert,et al.  Myeloproliferative neoplasm animal models. , 2012, Hematology/oncology clinics of North America.

[10]  O. Abdel-Wahab,et al.  The role of mutations in epigenetic regulators in myeloid malignancies , 2012, Nature Reviews Cancer.

[11]  B. Ebert,et al.  Distinct roles for long-term hematopoietic stem cells and erythroid precursor cells in a murine model of Jak2V617F-mediated polycythemia vera. , 2012, Blood.

[12]  M. Gönen,et al.  Genetic analysis of patients with leukemic transformation of myeloproliferative neoplasms shows recurrent SRSF2 mutations that are associated with adverse outcome. , 2012, Blood.

[13]  Ken Chen,et al.  Clonal architecture of secondary acute myeloid leukemia. , 2012, The New England journal of medicine.

[14]  W. Vainchenker,et al.  New mutations and pathogenesis of myeloproliferative neoplasms. , 2011, Blood.

[15]  O. Abdel-Wahab,et al.  Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. , 2011, Cancer cell.

[16]  D. Kent,et al.  Mouse models of myeloproliferative neoplasms: JAK of all grades , 2011, Disease Models & Mechanisms.

[17]  J. Spivak,et al.  Disease burden at the progenitor level is a feature of primary myelofibrosis: a multivariable analysis of 164 JAK2 V617F-positive myeloproliferative neoplasm patients. , 2011, Experimental hematology.

[18]  Erin F. Simonds,et al.  Novel mutations in the inhibitory adaptor protein LNK drive JAK-STAT signaling in patients with myeloproliferative neoplasms. , 2010, Blood.

[19]  Michael G. Kharas,et al.  Physiological Jak2V617F expression causes a lethal myeloproliferative neoplasm with differential effects on hematopoietic stem and progenitor cells. , 2010, Cancer cell.

[20]  R. Kusec,et al.  Two routes to leukemic transformation after a JAK2 mutation-positive myeloproliferative neoplasm. , 2010, Blood.

[21]  A. Tichelli,et al.  Clonal analysis of TET2 and JAK2 mutations suggests that TET2 can be a late event in the progression of myeloproliferative neoplasms. , 2010, Blood.

[22]  O. Abdel-Wahab,et al.  Genetic analysis of transforming events that convert chronic myeloproliferative neoplasms to leukemias. , 2010, Cancer research.

[23]  R. Levine,et al.  Mutation in TET2 in myeloid cancers. , 2009, The New England journal of medicine.

[24]  D. Gilliland,et al.  Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. , 2009, Blood.

[25]  A. Migliaccio,et al.  Pathological interactions between hematopoietic stem cells and their niche revealed by mouse models of primary myelofibrosis , 2009, Expert review of hematology.

[26]  D. Gilliland,et al.  TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis , 2009, Leukemia.

[27]  A. Hall,et al.  Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. , 2008, Blood.

[28]  E. Vellenga,et al.  Maximal STAT5-Induced Proliferation and Self-Renewal at Intermediate STAT5 Activity Levels , 2008, Molecular and Cellular Biology.

[29]  David Bryder,et al.  Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. , 2007, Cell stem cell.

[30]  M. Stratton,et al.  JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. , 2007, The New England journal of medicine.

[31]  Soon-Siong Teo,et al.  Leukemic blasts in transformed JAK2-V617F-positive myeloproliferative disorders are frequently negative for the JAK2-V617F mutation. , 2006, Blood.

[32]  P. Campbell,et al.  Mutation of JAK2 in the myeloproliferative disorders: timing, clonality studies, cytogenetic associations, and role in leukemic transformation. , 2006, Blood.

[33]  T. Golub,et al.  Transformation from committed progenitor to leukaemia stem cell initiated by MLL–AF9 , 2006, Nature.

[34]  Jill P. Mesirov,et al.  Comparative gene marker selection suite , 2006, Bioinform..

[35]  Sandra A. Moore,et al.  MPLW515L Is a Novel Somatic Activating Mutation in Myelofibrosis with Myeloid Metaplasia , 2006, PLoS medicine.

[36]  Pablo Tamayo,et al.  Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles , 2005, Proceedings of the National Academy of Sciences of the United States of America.

[37]  Stefan N Constantinescu,et al.  A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. , 2005, Nature.

[38]  Mario Cazzola,et al.  A gain-of-function mutation of JAK2 in myeloproliferative disorders. , 2005, The New England journal of medicine.

[39]  Sandra A. Moore,et al.  Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. , 2005, Cancer cell.

[40]  P. Campbell,et al.  Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders , 2005, The Lancet.

[41]  D. Charnock-Jones,et al.  vavCre Transgenic mice: A tool for mutagenesis in hematopoietic and endothelial lineages , 2002, Genesis.