Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
2.
JCI Insight ; 5(1)2020 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-31941838

RESUMEN

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription-quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options.


Asunto(s)
Proteínas de Unión al ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , MicroARNs/metabolismo , MicroARNs/farmacología , Mielofibrosis Primaria/tratamiento farmacológico , Proteínas Proto-Oncogénicas/efectos de los fármacos , Animales , Citocinas/metabolismo , Proteínas de Unión al ADN/genética , Dioxigenasas , Modelos Animales de Enfermedad , Histonas , Humanos , Inhibidores de las Cinasas Janus/uso terapéutico , Quinasas Janus/metabolismo , Ratones , MicroARNs/genética , Oxigenasas de Función Mixta , Mutación , Trastornos Mieloproliferativos , Nitrilos , Mielofibrosis Primaria/genética , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/genética , Pirazoles/uso terapéutico , Pirimidinas , Factor de Transcripción STAT3 , Transcriptoma , Estados Unidos
3.
Stem Cell Investig ; 4: 48, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28607922

RESUMEN

Acute myeloid leukemia (AML) is a cancer of the myeloid lineage of blood cells. Although significant progress has been made in treating many types of cancers during recent years, AML remains a deadly disease with survival rate lagging behind other blood cancers. A combination of toxic chemotherapies has been the standard AML treatment for more than 40 years. With intensive efforts to define the pathogenesis of AML, novel therapeutic drugs targeting key molecular defects in AML are being developed. Mutated in nearly 30% of AML, FMS-like tyrosine kinase 3 (FLT3) represents one of the most attractive targets. FLT3 mutants resulted from either internal tandem duplication (ITD) or point mutations possess enhanced kinase activity and cause constitutive activation of signaling. To date, several small molecule inhibitors of FLT3 have been developed but their clinical efficacy is limited due to a lack of potency and the generation of drug resistance. Therefore, next-generation FLT3 inhibitors overcoming these limitations are urgently in need. This review focuses on the pathological role of mutant FLT3 in the development of AML, the current status of FLT3 inhibitor development, and mechanisms underlining the development of resistance to existing FLT3 inhibitors.

4.
JCI Insight ; 2(7): e90932, 2017 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-28405618

RESUMEN

Myelofibrosis (MF) is a bone marrow disorder characterized by clonal myeloproliferation, aberrant cytokine production, extramedullary hematopoiesis, and bone marrow fibrosis. Although somatic mutations in JAK2, MPL, and CALR have been identified in the pathogenesis of these diseases, inhibitors of the Jak2 pathway have not demonstrated efficacy in ameliorating MF in patients. TGF-ß family members are profibrotic cytokines and we observed significant TGF-ß1 isoform overexpression in a large cohort of primary MF patient samples. Significant overexpression of TGF-ß1 was also observed in murine clonal MPLW515L megakaryocytic cells. TGF-ß1 stimulated the deposition of excessive collagen by mesenchymal stromal cells (MSCs) by activating the TGF-ß receptor I kinase (ALK5)/Smad3 pathway. MSCs derived from MPLW515L mice demonstrated sustained overproduction of both collagen I and collagen III, effects that were abrogated by ALK5 inhibition in vitro and in vivo. Importantly, use of galunisertib, a clinically active ALK5 inhibitor, significantly improved MF in both MPLW515L and JAK2V617F mouse models. These data demonstrate the role of malignant hematopoietic stem cell (HSC)/TGF-ß/MSC axis in the pathogenesis of MF, and provide a preclinical rationale for ALK5 blockade as a therapeutic strategy in MF.


Asunto(s)
Janus Quinasa 2/metabolismo , Mielofibrosis Primaria/tratamiento farmacológico , Pirazoles/farmacología , Quinolinas/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Receptores de Trombopoyetina/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Médula Ósea/patología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colágeno/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Megacariocitos/metabolismo , Megacariocitos/patología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mielofibrosis Primaria/metabolismo , Transducción de Señal , Proteína smad3/metabolismo
5.
PLoS One ; 9(7): e99017, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25036984

RESUMEN

The JAK2V617F mutation is found in the majority of patients with myeloproliferative neoplasms (MPNs). Transgenic expression of the mutant gene causes MPN-like phenotypes in mice. We have produced JAK2V617F mice with p53 null background. Some of these mice developed acute erythroleukemia. From one of these mice, we derived a cell line designated J53Z1. J53Z1 cells were stained positive for surface markers CD71 and CD117 but negative for Sca-1, TER-119, CD11b, Gr-1, F4/80, CD11c, CD317, CD4, CD8a, CD3e, B220, CD19, CD41, CD42d, NK-1.1, and FceR1. Real time PCR analyses demonstrated expressions of erythropoietin receptor EpoR, GATA1, and GATA2 in these cells. J53Z1 cells grew rapidly in suspension culture containing fetal bovine serum with a doubling time of ∼18 hours. When transplanted into C57Bl/6 mice, J53Z1 cells induced acute erythroleukemia with massive infiltration of tumor cells in the spleen and liver. J53Z1 cells were responsive to stimulation with erythropoietin and stem cell factor and were selectively inhibited by JAK2 inhibitors which induced apoptosis of the cells. Together, J53Z1 cells belong to the erythroid lineage, and they may be useful for studying the role of JAK2V617F in proliferation and differentiation of erythroid cells and for identifying potential therapeutic drugs targeting JAK2.


Asunto(s)
Línea Celular Tumoral/enzimología , Janus Quinasa 2/genética , Leucemia Eritroblástica Aguda/patología , Mutación Missense , Proteínas de Neoplasias/genética , Mutación Puntual , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral/efectos de los fármacos , Línea Celular Tumoral/trasplante , Cruzamientos Genéticos , Ensayos de Selección de Medicamentos Antitumorales , Eritropoyesis/efectos de los fármacos , Perfilación de la Expresión Génica , Genes p53 , Factores de Crecimiento de Célula Hematopoyética/farmacología , Humanos , Hígado/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/biosíntesis , Inhibidores de Proteínas Quinasas/farmacología , Bazo/patología
6.
J Hematol Oncol ; 7: 25, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24646493

RESUMEN

BACKGROUND: Myeloproliferative neoplasms (MPNs) are blood malignancies manifested in increased production of red blood cells, white blood cells, and/or platelets. A major molecular lesion associated with the diseases is JAK2V617F, an activation mutation form of tyrosine kinase JAK2. Cardiovascular events represent the leading cause of morbidity and mortality associated MPNs, but the underlying mechanism is not well understood. METHODS: Previously, we generated JAK2V617F transgenic mice which displayed MPN-like phenotypes. In the present study, we further characterized these mice by analyzing the time course of MPN phenotype development and associated cardiac abnormalities. We performed detailed histochemical staining of cardiac sections. RESULTS: JAK2V617F transgenic mice developed cardiomegaly as a subsequent event of increased blood cell production during the course of MPN phenotype development. The cardiomegaly is manifested in increased ventricular wall thickness and enlarged cardiomyocytes. Trichrome and reticulin staining revealed extensive collagen fibrosis in the heart of JAK2V617F transgenic mice. Thrombosis in the coronary artery and inflammatory cell infiltration into cardiac muscle were also observed in JAK2V617F transgenic mice, and the latter event was accompanied by fibrosis. CONCLUSION: JAK2V617F-induced blood disorders have a major impact on heart function and lead to cardiac hypertrophy. JAK2V617F transgenic mice represent an excellent model system to study both hematological malignancies and cardiovascular diseases.


Asunto(s)
Cardiomegalia/enzimología , Neoplasias Hematológicas/enzimología , Janus Quinasa 2/genética , Trastornos Mieloproliferativos/enzimología , Animales , Cardiomegalia/tratamiento farmacológico , Cardiomegalia/genética , Cardiomegalia/patología , Femenino , Fibrosis/enzimología , Fibrosis/genética , Fibrosis/patología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología
7.
Exp Hematol Oncol ; 1(1): 15, 2012 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23210734

RESUMEN

BACKGROUND: JAK2V617F, a gain-of-function mutant form of tyrosine kinase JAK2, is found in the majority of patients with Ph- myeloproliferative neoplasms (MPNs), a group of chronic hematological diseases that often lead to acute leukemia. The current study is intended to find other gene mutations that collaborate with JAK2V617F to cause leukemic transformation. METHODS: Total RNA and genomic DNA were isolated from two JAK2V617F-positive cell lines, namely, erythroleukemic HEL and megakaryoblastic leukemic SET-2 cells. Candidate genes were amplified by PCR and further sequenced. RESULTS: Homozygous mutations of the TP53 gene which encodes tumor suppressor p53 were found in HEL and SET-2 cells. While HEL cells, which have homozygous JAK2V617F, contain a rare M133K p53 mutation, SET-2 cells, which have a heterozygous JAK2V617F mutation, contain a common R248W p53 alteration. Western blot analyses revealed high levels of p53 expression in both cells. M133K and R248W are located in the DNA binding domain of p53. Structural analyses revealed that they potentially disrupt the interaction of p53 with DNA, thereby causing loss of p53 function. CONCLUSIONS: JAK2V617F and p53 mutations coexist in leukemia cells. We believe that JAK2V617F is able to drive leukemic transformation when the function of tumor suppressor p53 is lost. The interplay of JAK2V617F with p53 may affect the progression of MPNs.

8.
J Hematol Oncol ; 5: 39, 2012 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-22800464

RESUMEN

BACKGROUND: Gain-of-function mutations of tyrosine kinase FLT3 are frequently found in acute myeloid leukemia (AML). This has made FLT3 an important marker for disease diagnosis and a highly attractive target for therapeutic drug development. This study is intended to generate a sensitive substrate for assays of the FLT3 enzymatic activity. METHODS: We expressed in Escherichia coli cells a glutathione S-transferase (GST) fusion protein designated GST-FLT3S, which contains a peptide sequence derived from an autophosphorylation site of FLT3. The protein was used to analyze tyrosine kinase activity of baculovirus-expressed FLT3 and crude cell extracts of bone marrow cells from AML patients. It was also employed to perform FLT3 kinase assays for FLT3 inhibitor screening. RESULTS: GST-FLT3S in solution or on beads was strongly phosphorylated by recombinant proteins carrying the catalytic domain of wild type FLT3 and FLT3D835 mutants, with the latter exhibiting much higher activity and efficiency. GST-FLT3S was also able to detect elevated tyrosine kinase activity in bone marrow cell extracts from AML patients. A small-scale inhibitor screening led to identification of several potent inhibitors of wild type and mutant forms of FLT3. CONCLUSIONS: GST-FLT3S is a sensitive protein substrate for FLT3 assays. It may find applications in diagnosis of diseases related to abnormal FLT3 activity and in inhibitor screening for drug development.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Leucemia Mieloide Aguda/metabolismo , Fragmentos de Péptidos/farmacología , Proteínas Tirosina Quinasas/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Tirosina Quinasa 3 Similar a fms/metabolismo , Médula Ósea/metabolismo , Médula Ósea/patología , Evaluación Preclínica de Medicamentos , Glutatión Transferasa/genética , Glutatión Transferasa/metabolismo , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Mutación/genética , Fosforilación/efectos de los fármacos , Proteínas Recombinantes de Fusión/antagonistas & inhibidores , Proteínas Recombinantes de Fusión/genética , Especificidad por Sustrato , Células Tumorales Cultivadas , Tirosina Quinasa 3 Similar a fms/antagonistas & inhibidores , Tirosina Quinasa 3 Similar a fms/genética
9.
J Cell Biochem ; 104(5): 1843-52, 2008 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-18393358

RESUMEN

The myotubularin (MTM) enzymes are phosphatidylinositol 3-phosphate (PI3P) and phosphatidylinositol 3,5-bisphosphate phosphatases. Mutation of MTM1, the founder member of this family, is responsible for X-linked myotubular myopathy in humans. Here, we have isolated and characterized a Caenorhabditis elegans homology of the enzymes designated ceMTM3. ceMTM3 preferably dephosphorylates PI3P and contains a FYVE lipid-binding domain at its C-terminus which binds PI3P. Immunoblotting analyses revealed that the enzyme is expressed during the early development and adulthood of the animal. Immunofluorescent staining revealed predominant expression of the enzyme in eggs and muscles. Knockdown of the enzyme by using feeding-based RNA interference resulted in an increased level of PI3P and caused severe impairment of body movement of the worms at their post-reproductive ages and significantly shortened their lifespan. This study thus reveals an important role of the MTM phosphatases in maintaining muscle function, which may have clinical implications in prevention and treatment of sarcopenia.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/enzimología , Proteínas Tirosina Fosfatasas no Receptoras/química , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Animales , Clonación Molecular , Locomoción , Fosfatos de Fosfatidilinositol/metabolismo , Estructura Terciaria de Proteína , Transporte de Proteínas , Interferencia de ARN , Especificidad por Sustrato , Análisis de Supervivencia
10.
Exp Hematol ; 35(11): 1624-32, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17764811

RESUMEN

OBJECTIVE: Identification of JAK2V617F in myeloproliferative disorders makes JAK2 an important marker for disease diagnosis and a highly attractive target for therapeutic drug development. This study is intended to identify a sensitive and specific substrate for assays of the JAK2 enzymatic activity. METHODS: We expressed a glutathione S-transferase (GST) fusion protein designated GST-JAKS, which carries a peptide sequence derived from the autophosphorylation sites of human JAK2. The protein was purified from Escherichia coli cells and was used to analyze to tyrosine kinase activities of purified enzymes and crude cell extracts from cells, including mononuclear cells of JAK2V617F -positive polycythemia vera blood. It was also used to perform JAK2 kinase assays to screen inhibitors of JAK2. RESULTS: GST-JAKS is strongly phosphorylated by activated forms of JAK2 including JAK2V617F and recombinant protein containing its catalytic domain alone. It showed minimal responses to wild-type JAK2 and was not phosphorylated by the epidermal growth receptor and the insulin receptor tyrosine kinases. Kinase assays with GST-JAKS provide a sharp contrast between wild-type and mutant JAK2,V617F and are sensitive enough to detect minute amounts of JAK2V617F found in crude cell extracts. Assays can be scaled up to screen for inhibitors of JAK2 in a dot blot format. CONCLUSION: GST-JAKS is sensitive and specific protein substrate for JAK2 assays. It may have clinical applications in diagnosis of diseases related to abnormal JAK2 activity. It is also an excellent substrate for development of large scale assays to screen JAK2 inhibitors.


Asunto(s)
Pruebas Enzimáticas Clínicas/métodos , Janus Quinasa 2/análisis , Janus Quinasa 2/metabolismo , Mutación Missense , Proteínas Recombinantes de Fusión/metabolismo , Evaluación Preclínica de Medicamentos/métodos , Inhibidores Enzimáticos , Glutatión Transferasa/genética , Humanos , Janus Quinasa 2/genética , Quinasas Janus/genética , Quinasas Janus/metabolismo , Fosforilación , Especificidad por Sustrato
11.
J Biol Chem ; 282(6): 3428-32, 2007 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-17178722

RESUMEN

JAK2(V617F), a mutant of tyrosine kinase JAK2, is found in most patients with polycythemia vera (PV) and a substantial proportion of patients with idiopathic myelofibrosis or essential thrombocythemia. The JAK2 mutant displays a much increased kinase activity and generates a PV-like phenotype in mouse bone marrow transplant models. This study shows that the anti-cancer drug erlotinib (Tarceva) is a potent inhibitor of JAK2(V617F) activity. In vitro colony culture assays revealed that erlotinib at micro-molar concentrations effectively suppresses the growth and expansion of PV hematopoietic progenitor cells while having little effect on normal cells. Furthermore, JAK2(V617F)-positive cells from PV patients show greater susceptibility to the inhibitor than their negative counterparts. Similar inhibitory effects were found with the JAK2(V617F)-positive human erythroleukemia HEL cell line. These data suggest that erlotinib may be used for treatment of JAK2(V617F)-positive PV and other myeloproliferative disorders.


Asunto(s)
Sustitución de Aminoácidos/genética , Proliferación Celular/efectos de los fármacos , Inhibidores de Crecimiento/farmacología , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/metabolismo , Policitemia Vera/enzimología , Policitemia Vera/patología , Quinazolinas/farmacología , Animales , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Clorhidrato de Erlotinib , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/enzimología , Células Madre Hematopoyéticas/patología , Humanos , Células Jurkat , Ratones , Fenilalanina/genética , Policitemia Vera/tratamiento farmacológico , Policitemia Vera/genética , Inhibidores de Proteínas Quinasas/farmacología , Valina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...