Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Haematologica ; 101(10): 1190-1199, 2016 10.
Article in English | MEDLINE | ID: mdl-27418650

ABSTRACT

Juvenile myelomonocytic leukemia is a rare myeloproliferative neoplasm characterized by hyperactive RAS signaling. Neurofibromin1 (encoded by the NF1 gene) is a negative regulator of RAS activation. Patients with neurofibromatosis type 1 harbor loss-of-function mutations in NF1 and have a 200- to 500-fold increased risk of juvenile myelomonocytic leukemia. Leukemia cells from patients with juvenile myelomonocytic leukemia display hypersensitivity to certain cytokines, such as granulocyte-macrophage colony-stimulating factor. The granulocyte-macrophage colony-stimulating factor receptor utilizes pre-associated JAK2 to initiate signals after ligand binding. JAK2 subsequently activates STAT5, among other downstream effectors. Although STAT5 is gaining recognition as an important mediator of growth factor signaling in myeloid leukemias, the contribution of STAT5 to the development of hyperactive RAS-initiated myeloproliferative disease has not been well described. In this study, we investigated the consequence of STAT5 attenuation via genetic and pharmacological approaches in Nf1-deficient murine models of juvenile myelomonocytic leukemia. We found that homozygous Stat5 deficiency extended the lifespan of Nf1-deficient mice and eliminated the development of myeloproliferative neoplasm associated with Nf1 gene loss. Likewise, we found that JAK inhibition with ruxolitinib attenuated myeloproliferative neoplasm in Nf1-deficient mice. Finally, we found that primary cells from a patient with KRAS-mutant juvenile myelomonocytic leukemia displayed reduced colony formation in response to JAK2 inhibition. Our findings establish a central role for STAT5 activation in the pathogenesis of juvenile myelomonocytic leukemia and suggest that targeting this pathway may be of clinical utility in these patients.


Subject(s)
Janus Kinase 2/metabolism , Leukemia, Myelomonocytic, Juvenile/etiology , Myeloproliferative Disorders/etiology , Neurofibromin 1/deficiency , STAT5 Transcription Factor/physiology , Animals , Disease Models, Animal , Humans , Leukemia, Myeloid/etiology , Leukemia, Myeloid/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Mice , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction
2.
Am J Hematol ; 86(7): 579-85, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21681782

ABSTRACT

Juvenile Myelomonocytic Leukemia (JMML) is a relentlessly progressive myeloproliferative/myelodysplastic (MPD/MDS) hematopoietic disorder more common in patients with any one of at least three distinct genetic lesions, specifically NF1 gene loss and PTPN11 and NRAS mutations. NF1 and PTPN11 are molecular lesions associated with Neurofibromatosis Syndrome Type I (NF1 Syndrome) and Noonan's Syndrome, respectively. The occurrence of JMML is rare; even among those predisposed with these syndromes to development of disease, and secondary genetic events likely contribute to the development and progression of disease. In NF1 syndrome, loss of p53 function is a common event in solid tumors, but uncommon in JMML, suggesting that the p53 pathway may be modified by other events in this hematopoietic disorder. The work presented here investigates the possible role of the p19(Arf) (p19) tumor suppressor in development of MPD associated with Nf1 gene loss in mice. We find that Nf1 mutant hematopoietic cells with loss of p19 develop accelerated hematopoietic disease similar to acute leukemia with a variable phenotype. This suggests that p19 may play a role in development of JMML and evaluation of the human p19 homolog (p14(ARF)) in JMML may be informative.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Leukemia, Myelomonocytic, Juvenile/genetics , Neurofibromin 1/genetics , Acute Disease , Animals , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Humans , Leukemia, Myelomonocytic, Juvenile/metabolism , Leukemia, Myelomonocytic, Juvenile/pathology , Mice , Mice, Mutant Strains , Neurofibromatosis 1/genetics , Neurofibromatosis 1/metabolism , Neurofibromatosis 1/pathology , Neurofibromin 1/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
3.
Leuk Res ; 31(8): 1107-13, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17222906

ABSTRACT

Inactivation of the NF1 tumor suppressor causes myeloproliferative diseases. NF1 encodes a GTPase activating protein (GAP) for Ras. Myeloid cells with loss of NF1 have high levels of Ras-GTP, functionally equivalent to the effects of RAS oncogenes. We investigated the effects of the NF1 GAP-related domain (GRD) in proliferation, apoptosis and Ras-GTP levels in Nf1-negative acute myeloid leukemia (AML) cells. In AML cells, with cooperating mutations, the expression of the neurofibromin GRD causes significant reductions of N- and K-Ras-GTP levels, which is not incompatible with AML cell survival, but which is strongly selected against due to suppression of proliferation.


Subject(s)
Cell Proliferation , Down-Regulation , GTPase-Activating Proteins/physiology , Leukemia, Myeloid, Acute/pathology , Neurofibromin 1/physiology , Proto-Oncogene Proteins p21(ras)/metabolism , Animals , Blotting, Western , Guanosine Triphosphate/metabolism , Humans , Leukemia, Myeloid, Acute/metabolism , Mice , Mice, SCID , Protein Structure, Tertiary , Tumor Cells, Cultured
4.
Blood ; 109(4): 1687-91, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17090653

ABSTRACT

Neurofibromatosis type 1 (NF1) syndrome is caused by germline mutations in the NF1 tumor suppressor, which encodes neurofibromin, a GTPase activating protein for Ras. Children with NF1 are predisposed to juvenile myelomonocytic leukemia (JMML) and lethally irradiated mice given transplants with homozygous Nf1 mutant (Nf1-/-) hematopoietic stem cells develop a fatal myeloproliferative disorder (MPD) that models JMML. We investigated the requirement for signaling through the GM-CSF receptor to initiate and sustain this MPD by generating Nf1 mutant hematopoietic cells lacking the common beta chain (Beta c) of the GM-CSF receptor. Mice reconstituted with Nf1-/-, beta c-/- stem cells did not develop evidence of MPD despite the presence of increased number of immature hematopoietic progenitors in the bone marrow. Interestingly, when the Mx1-Cre transgene was used to inactivate a conditional Nf1 mutant allele in hematopoietic cells, concomitant loss of beta c-/- reduced the severity of the MPD, but did not abrogate it. Whereas inhibiting GM-CSF signaling may be of therapeutic benefit in JMML, our data also demonstrate aberrant proliferation of Nf1-/-myeloid progenitors that is independent of signaling through the GM-CSF receptor.


Subject(s)
Leukemia, Myelomonocytic, Chronic/etiology , Myeloproliferative Disorders/etiology , Neurofibromatosis 1/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Animals , Disease Models, Animal , Hematopoietic Stem Cell Transplantation , Leukemia, Myelomonocytic, Chronic/prevention & control , Mice , Mice, Mutant Strains , Myeloproliferative Disorders/prevention & control , Signal Transduction
5.
Blood ; 106(3): 1054-62, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15831708

ABSTRACT

To create a model in which to study the effects of RAS dysregulation in hematopoietic disease, we developed separate founder lines of transgenic mice, with the tetracycline transactivator (tTA) driven by the Vav hematopoietic promoter in one line and NRASV12 driven by the tetracycline responsive element (TRE2) in the other. When these lines are crossed, doubly transgenic animals uniformly develop a disease similar to human aggressive systemic mastocytosis (ASM) or mast cell leukemia (MCL) when they are between 2 and 4 months of age. Disease is characterized by tissue infiltrates of large, well-differentiated mast cells in the spleen, liver, skin, lung, and thymus. Analysis of bone sections shows small to large foci of similarly well-differentiated mast cells. Results also show that transgene expression and diseases are repressible through the administration of doxycycline in the drinking water of affected animals, indicating that NRASV12 expression is required to initiate and maintain disease in doubly transgenic mice. Our inducible system of transgenes, developed as a model of mutant NRASV12 oncogene-driven myeloid disease, will be useful for studying the role of RAS dysregulation in hematopoietic disease in general and in discrete human diseases, specifically ASM and MCL.


Subject(s)
Disease Models, Animal , Gene Expression Regulation , Genes, ras , Mastocytosis/genetics , Animals , Doxycycline/pharmacology , Leukemia, Mast-Cell , Leukemic Infiltration , Mastocytosis/etiology , Mastocytosis/pathology , Mastocytosis, Systemic , Mice , Mice, Transgenic , Response Elements , Tetracycline , Transduction, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...