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.
PLoS One ; 8(9): e75472, 2013.
Article in English | MEDLINE | ID: mdl-24086539

ABSTRACT

Utilizing ENU mutagenesis, we identified a mutant mouse with elevated platelets. Genetic mapping localized the mutation to an interval on chromosome 19 that encodes the Jak2 tyrosine kinase. We identified a A3056T mutation resulting in a premature stop codon within exon 19 of Jak2 (Jak2(K915X)), resulting in a protein truncation and functionally inactive enzyme. This novel platelet phenotype was also observed in mice bearing a hemizygous targeted disruption of the Jak2 locus (Jak2(+/-)). Timed pregnancy experiments revealed that Jak2(K915X/K915X) and Jak2(-/-) displayed embryonic lethality; however, Jak2(K915X/K915X) embryos were viable an additional two days compared to Jak2(-/-) embryos. Our data suggest that perturbing JAK2 activation may have unexpected consequences in elevation of platelet number and correspondingly, important implications for treatment of hematological disorders with constitutive Jak2 activity.


Subject(s)
Blood Platelets/cytology , Janus Kinase 2/genetics , Phenotype , Animals , Blotting, Western , Chromosome Mapping , Ethylnitrosourea , Fluorouracil , Genotype , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mutagenesis/genetics , Phenylhydrazines , Point Mutation/genetics , Sequence Analysis, DNA
2.
Exp Hematol ; 40(1): 48-60, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21924221

ABSTRACT

The ability of random mutagenesis techniques to annotate the mammalian genome can be hampered due to genetic redundancy and compensatory pathways that mask heterozygous mutations under homeostatic conditions. The objective of this study was to devise a pharmacologically sensitized screen using the chemotherapeutic drug, 5-fluorouracil (5FU), to induce cytopenia. 5FU dose was optimized in the 129/SvImJ, C57BL/6J, BALB/cJ, and C3H/HeJ strains of laboratory mice. N-ethyl-N-nitrosourea (ENU) mutagenesis was performed on 129/SvImJ males and phenotypic variants were identified by backcrossing on to the C57BL/6J background. G1 animals were challenged with 100 µg/g 5FU and phenodeviants with altered platelet recovery were monitored. Of 546 G1 animals tested, 15 phenodeviants were identified that displayed increased baseline platelet number, a platelet overshoot, or delayed platelet recovery, thereby demonstrating the utility of this approach for uncovering mutations in megakaryocyte and platelet development. Four G1 mice were selected for further analysis. The phenotypes were heritable in all four strains and genetic mapping identified a chromosome location in two of the three G2 lines tested. In conclusion, our group has developed a sensitized random mutagenesis screen utilizing 5FU and has shown that the strain combination of 129/SvImJ × C57BL/6J is robust for identification of founder lines with defects in megakaryocyte and platelet development.


Subject(s)
Antineoplastic Agents/pharmacology , Ethylnitrosourea/pharmacology , Fluorouracil/pharmacology , Mutagenesis/drug effects , Mutation/genetics , Thrombocytopenia/chemically induced , Thrombopoiesis/genetics , Animals , Female , Male , Mice , Mice, 129 Strain , Mice, Congenic , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mutagenesis/genetics , Thrombopoiesis/drug effects
3.
Blood ; 116(3): 428-36, 2010 Jul 22.
Article in English | MEDLINE | ID: mdl-20445019

ABSTRACT

The activation of Fli-1, an Ets transcription factor, is the critical genetic event in Friend murine leukemia virus (F-MuLV)-induced erythroleukemia. Fli-1 overexpression leads to erythropoietin-dependent erythroblast proliferation, enhanced survival, and inhibition of terminal differentiation, through activation of the Ras pathway. However, the mechanism by which Fli-1 activates this signal transduction pathway has yet to be identified. Down-regulation of the Src homology 2 (SH2) domain-containing inositol-5-phosphatase-1 (SHIP-1) is associated with erythropoietin-stimulated erythroleukemic cells and correlates with increased proliferation of transformed cells. In this study, we have shown that F-MuLV-infected SHIP-1 knockout mice display accelerated erythroleukemia progression. In addition, RNA interference (RNAi)-mediated suppression of SHIP-1 in erythroleukemia cells activates the phosphatidylinositol 3-kinase (PI 3-K) and extracellular signal-regulated kinase/mitogen-activated protein kinase (ERK/MAPK) pathways, blocks erythroid differentiation, accelerates erythropoietin-induced proliferation, and leads to PI 3-K-dependent Fli-1 up-regulation. Chromatin immunoprecipitation and luciferase assays confirmed that Fli-1 binds directly to an Ets DNA binding site within the SHIP-1 promoter and suppresses SHIP-1 transcription. These data provide evidence to suggest that SHIP-1 is a direct Fli-1 target, SHIP-1 and Fli-1 regulate each other in a negative feedback loop, and the suppression of SHIP-1 by Fli-1 plays an important role in the transformation of erythroid progenitors by F-MuLV.


Subject(s)
Leukemia, Erythroblastic, Acute/etiology , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Protein c-fli-1/metabolism , Animals , Base Sequence , Binding Sites/genetics , Cell Line , DNA/genetics , DNA/metabolism , DNA Primers/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , Friend murine leukemia virus/pathogenicity , Humans , Inositol Polyphosphate 5-Phosphatases , Leukemia, Erythroblastic, Acute/genetics , Leukemia, Erythroblastic, Acute/virology , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , Molecular Sequence Data , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Phosphorylation , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics
4.
J Allergy Clin Immunol ; 119(1): 123-31, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17208593

ABSTRACT

BACKGROUND: Src homology 2 domain-containing inositol 5-phosphatase 1 (SHIP-1) controls the intracellular level of the phosphoinositide 3-kinase product phosphotidylinositol-3,4,5-trisphosphate and functions as a negative regulator of cytokine and immune receptor signaling. Emerging evidence suggests that the phosphoinositide 3-kinase pathway might be involved in allergic inflammation in the lung. However, the functional relevance of SHIP-1 in the T(H)2 activation pathway has not been established. SHIP-1(-/-) mice have spontaneous myeloproliferative inflammation in the lung, the nature of which has not been elucidated. We hypothesized that SHIP-1 plays an important role as a regulator in pulmonary allergic inflammation and in maintaining lung homeostasis. OBJECTIVE: To test our hypothesis, we characterized the pulmonary phenotype of SHIP-1(-/-) mice. RESULTS: Analyses of lung histopathology and bronchoalveolar lavage cellularity revealed that the majority of SHIP-1(-/-) mice had progressive and severe pulmonary inflammation of macrophages, lymphocytes, neutrophils, and eosinophils; mucous hyperplasia; airway epithelial hypertrophy; and subepithelial fibrosis. These pathologic changes were accompanied by exaggerated production of T(H)2 cytokines and chemokines, including IL-4, IL-13, eotaxin, and monocyte chemoattractant protein 1, in the lung. Furthermore, the number of mast cells significantly increased, and many of these cells were undergoing degranulation, which was correlated with increased content and spontaneous release of histamine in the lung tissue of SHIP-1(-/-) mice. CONCLUSION: These findings provide strong evidence that mice lacking SHIP-1 have an allergic inflammation in the lung, suggesting that SHIP-1 plays an important role in regulating the T(H)2 signaling pathway and in maintaining lung homeostasis. CLINICAL IMPLICATIONS: SHIP-1 as a regulator might be a potential therapeutic target for controlling allergic inflammation in diseases such as asthma.


Subject(s)
Inflammation/immunology , Phosphoric Monoester Hydrolases/deficiency , Respiratory Hypersensitivity/immunology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Chitinases/metabolism , Cytokines/biosynthesis , Cytokines/genetics , Histamine/metabolism , Immunoglobulin E/analysis , Inflammation/metabolism , Inositol Polyphosphate 5-Phosphatases , Leukocyte Count , Lung/immunology , Lung/metabolism , Lung/pathology , Mice , Mice, Knockout , Mucin 5AC , Mucins/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , RNA, Messenger/biosynthesis , Respiratory Hypersensitivity/metabolism , Th2 Cells/immunology , Up-Regulation
5.
Blood ; 105(2): 552-61, 2005 Jan 15.
Article in English | MEDLINE | ID: mdl-15213094

ABSTRACT

Erythropoietin (EPO) activates many distinct signal transduction cascades on engagement of its receptor. Deletion of the EPO, EPO receptor (EPO-R), or JAK2 genes in mice results in embryonic lethality due to a fatal anemia. EPO activates signal transducer and activator of transcription 1 (STAT1), STAT3, and STAT5a/b transcription factors in erythroid cell lines. Studies have focused on STAT5 as the primary target of EPO-dependent JAK2 activation. However, STAT5a/b(-/-) mice are viable, displaying a nonfatal anemia during embryogenesis, and delayed differentiation in adult erythropoiesis. Importantly, EPO-R cytoplasmic tyrosines are dispensable for viability in vivo. Interestingly, no cytoplasmic tyrosines are required for phosphorylation of STAT1. This led us to examine whether STAT1-deficient mice have altered erythropoiesis. A shift in erythropoiesis was observed in STAT1(-/-) mice, with reduced bone marrow-derived erythroid colony-forming units (CFU-Es) and a compensatory increase in splenic burst-forming units (BFU-Es) and CFU-Es. Both types of splenic-derived cells displayed EPO hyperresponsiveness. A 1.6-fold reduction in total CFU-Es was observed in STAT1-deficient mice, whereas total BFU-Es were comparable. Flow cytometry of STAT1-deficient erythroid cells revealed a less differentiated phenotype, associated with increased apoptosis of early erythroblasts. STAT1-deficient erythroblasts from phenylhydrazine-primed mice displayed enhanced phosphorylation of STAT5a/b, Erk1/2, and protein kinase B (PKB)/Akt. These results illustrate that STAT1 plays an important role in the regulation of erythropoiesis.


Subject(s)
DNA-Binding Proteins/genetics , Erythroid Precursor Cells/physiology , Erythropoiesis/physiology , Trans-Activators/genetics , Anemia/physiopathology , Animals , Apoptosis/physiology , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation/physiology , Cell Division/physiology , DNA-Binding Proteins/metabolism , Erythroblasts/enzymology , Erythroid Precursor Cells/cytology , Erythropoietin/pharmacology , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Mutant Strains , Milk Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , STAT1 Transcription Factor , STAT5 Transcription Factor , Signal Transduction/physiology , Spleen/cytology , Trans-Activators/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...