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











Base de dados
Intervalo de ano de publicação
1.
Blood ; 124(13): 2104-15, 2014 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-25143485

RESUMO

Megakaryopoiesis is a 2-step differentiation process, regulated by thrombopoietin (TPO), on binding to its cognate receptor myeloproliferative leukemia (MPL). This receptor associates with intracytoplasmic tyrosine kinases, essentially janus kinase 2 (JAK2), which regulates MPL stability and cell-surface expression, and mediates TPO-induced signal transduction. We demonstrate that JAK2 and MPL mediate TPO-induced proliferation arrest and megakaryocytic differentiation of the human megakaryoblastic leukemia cell line UT7-MPL. A decrease in JAK2 or MPL protein expression, and JAK2 chemical inhibition, suppress this antiproliferative action of TPO. The expression of JAK2 and MPL, which progressively increases along normal human megakaryopoiesis, is decreased in platelets of patients diagnosed with JAK2- or MPL-mutated essential thrombocytemia and primary myelofibrosis, 2 myeloproliferative neoplasms in which megakaryocytes (MKs) proliferate excessively. Finally, low doses of JAK2 chemical inhibitors are shown to induce a paradoxical increase in MK production, both in vitro and in vivo. We propose that JAK2 and MPL expression levels regulate megakaryocytic proliferation vs differentiation in both normal and pathological conditions, and that JAK2 chemical inhibitors could promote a paradoxical thrombocytosis when used at suboptimal doses.


Assuntos
Autoantígenos/metabolismo , Diferenciação Celular , Iodeto Peroxidase/metabolismo , Proteínas de Ligação ao Ferro/metabolismo , Janus Quinase 2/metabolismo , Megacariócitos/citologia , Megacariócitos/metabolismo , Receptores de Trombopoetina/metabolismo , Animais , Autoantígenos/genética , Plaquetas/metabolismo , Pontos de Checagem do Ciclo Celular/genética , Diferenciação Celular/genética , Linhagem Celular , Proliferação de Células , Expressão Gênica , Humanos , Iodeto Peroxidase/genética , Proteínas de Ligação ao Ferro/genética , Janus Quinase 2/genética , Camundongos , Fenótipo , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , RNA Interferente Pequeno/genética , Receptores de Trombopoetina/genética , Trombocitemia Essencial/genética , Trombocitemia Essencial/metabolismo
3.
Cancer Res ; 71(8): 2901-15, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21487043

RESUMO

Primary myelofibrosis (PMF) is characterized by increased number of hematopoietic progenitors and a dysmegakaryopoiesis which supports the stromal reaction defining this disease. We showed that increased ligand (FL) levels in plasma, hematopoietic progenitors, and stromal cells from PMF patients were associated with upregulation of the cognate Flt3 receptor on megakaryocytic (MK) cells. This connection prompted us to study a functional role for the FL/Flt3 couple in PMF dysmegakaryopoiesis, as a route to reveal insights into pathobiology and therapy in this disease. Analysis of PMF CD34(+) and MK cell transcriptomes revealed deregulation of the mitogen-activated protein kinase (MAPK) pathway along with Flt3 expression. In PMF patients, a higher proportion of circulating Flt3(+)CD34(+)CD41(+) cells exhibited an increased MAPK effector phosphorylation independently of Jak2(V617F) mutation. Activation of FL/Flt3 axis in PMF MK cell cultures, in response to FL, induced activation of the p38-MAPK cascade, which is known to be involved in inflammation, also increasing expression of its target genes (NFATC4, p53, AP-1, IL-8). Inhibiting Flt3 or MAPK or especially p38 by chemical, antibody, or silencing strategies restored megakaryopoiesis and reduced phosphorylation of Flt3 and p38 pathway effectors, confirming the involvement of Flt3 in PMF dysmegakaryopoiesis via p38 activation. In addition, in contrast to healthy donors, MK cells derived from PMF CD34(+) cells exhibited an FL-induced migration that could be reversed by p38 inhibition. Taken together, our results implicate the FL/Flt3 ligand-receptor complex in PMF dysmegakaryopoiesis through persistent p38-MAPK activation, with implications for therapeutic prospects to correct altered megakaryopoiesis in an inflammatory context.


Assuntos
Megacariócitos/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mielofibrose Primária/metabolismo , Tirosina Quinase 3 Semelhante a fms/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Antígenos CD34/biossíntese , Ativação Enzimática , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Leucócitos Mononucleares/enzimologia , Leucócitos Mononucleares/metabolismo , Leucócitos Mononucleares/patologia , Sistema de Sinalização das MAP Quinases , Megacariócitos/enzimologia , Megacariócitos/patologia , Proteínas de Membrana/sangue , Proteínas de Membrana/metabolismo , Proteínas de Membrana/farmacologia , Fosforilação , Mielofibrose Primária/sangue , Mielofibrose Primária/enzimologia , Células Estromais/enzimologia , Células Estromais/metabolismo , Células Estromais/patologia , Tirosina Quinase 3 Semelhante a fms/biossíntese
4.
PLoS Biol ; 8(9)2010 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-20838657

RESUMO

Thrombopoietin (TPO) via signaling through its cognate receptor MPL is a key cytokine involved in the regulation of megakaryocyte differentiation leading to platelet production. Mature megakaryocytes are polyploid cells that have arrested DNA replication and cellular proliferation but continue sustained protein synthesis. Here, we show that TPO induces cell-cycle arrest in the megakaryocytic UT7-MPL cell line by the activation of the ERK/MAPK pathway, induction of p21CIP transcription, and senescence markers through EGR1 activation. A similar senescence-like process was also detected in normal primary postmitotic megakaryocytes. In contrast, senescence was not observed in malignant megakaryocytes derived from primary myelofibrosis patients (a form of chronic myeloid hemopathy). Our data indicate that polyploid mature megakaryocytes receive signals from TPO to arrest cell proliferation and enter a senescent-like state. An escape from this physiological process may be associated with certain myeloproliferative neoplasms leading to abnormal megakaryocytic proliferation.


Assuntos
Ciclo Celular , Proliferação de Células , Senescência Celular , Megacariócitos/citologia , Linhagem Celular , Humanos , Megacariócitos/efeitos dos fármacos , Trombopoetina/farmacologia
5.
Blood ; 116(26): 5961-71, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20870899

RESUMO

Activating mutations in signaling molecules, such as JAK2-V617F, have been associated with myeloproliferative neoplasms (MPNs). Mice lacking the inhibitory adaptor protein Lnk display deregulation of thrombopoietin/thrombopoietin receptor signaling pathways and exhibit similar myeloproliferative characteristics to those found in MPN patients, suggesting a role for Lnk in the molecular pathogenesis of these diseases. Here, we showed that LNK levels are up-regulated and correlate with an increase in the JAK2-V617F mutant allele burden in MPN patients. Using megakaryocytic cells, we demonstrated that Lnk expression is regulated by the TPO-signaling pathway, thus indicating an important negative control loop in these cells. Analysis of platelets derived from MPN patients and megakaryocytic cell lines showed that Lnk can interact with JAK2-WT and V617F through its SH2 domain, but also through an unrevealed JAK2-binding site within its N-terminal region. In addition, the presence of the V617F mutation causes a tighter association with Lnk. Finally, we found that the expression level of the Lnk protein can modulate JAK2-V617F-dependent cell proliferation and that its different domains contribute to the inhibition of multilineage and megakaryocytic progenitor cell growth in vitro. Together, our results indicate that changes in Lnk expression and JAK2-V617F-binding regulate JAK2-mediated signals in MPNs.


Assuntos
Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação/genética , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Proteínas/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proliferação de Células , Células Cultivadas , Humanos , Immunoblotting , Imunoprecipitação , Peptídeos e Proteínas de Sinalização Intracelular , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Transtornos Mieloproliferativos/patologia , Ligação Proteica , Proteínas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombopoetina/metabolismo
6.
PLoS One ; 5(1): e8893, 2010 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-20126644

RESUMO

BACKGROUND: Myeloproliferative disorders are characterized by clonal expansion of normal mature blood cells. Acquired mutations giving rise to constitutive activation of the JAK2 tyrosine kinase has been shown to be present in the majority of patients. Since the demonstration that the V617F mutation in the exon 14 of the JAK2 gene is present in about 90% of patients with Polycythemia Vera (PV), the detection of this mutation has become a key tool for the diagnosis of these patients. More recently, additional mutations in the exon 12 of the JAK2 gene have been described in 5 to 10% of the patients with erythrocytosis. According to the updated WHO criteria the presence of these mutations should be looked for in PV patients with no JAK2 V617F mutation. Reliable and accurate methods dedicated to the detection of these highly variable mutations are therefore necessary. METHODS/FINDINGS: For these reasons we have defined the conditions of a High Resolution DNA Melting curve analysis (HRM) method able to detect JAK2 exon 12 mutations. After having validated that the method was able to detect mutated patients, we have verified that it gave reproducible results in repeated experiments, on DNA extracted from either total blood or purified granulocytes. This HRM assay was further validated using 8 samples bearing different mutant sequences in 4 different laboratories, on 3 different instruments. CONCLUSION: The assay we have developed is thus a valid method, adapted to routine detection of JAK2 exon 12 mutations with highly reproducible results.


Assuntos
Éxons , Janus Quinase 2/genética , Laboratórios/normas , Mutação , Policitemia Vera/genética , Sequência de Bases , Primers do DNA , Humanos , Reação em Cadeia da Polimerase
7.
Blood ; 111(10): 4922-9, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18250227

RESUMO

Myeloproliferative diseases (MPDs) represent the commonest cause of splanchnic vein thrombosis (SVT), including Budd-Chiari syndrome (BCS) and portal vein thrombosis (PVT), but their diagnosis is hampered by changes secondary to portal hypertension, while their influence in the outcome of SVT remains unclear. We assessed the diagnostic and prognostic value of JAK2 and MPL515 mutations in 241 SVT patients (104 BCS, 137 PVT). JAK2V617F was found in 45% of BCS and 34% of PVT, while JAK2 exon 12 and MPL515 mutations were not detected. JAK2V617F was found in 96.5% of patients with bone marrow (BM) changes specific for MPD and endogenous erythoid colonies, but also in 58% of those with only one feature and in 7% of those with neither feature. Stratifying MPD diagnosis first on JAK2V617F detection would have avoided BM investigations in 40% of the patients. In BCS, presence of MPD carried significantly poorer baseline prognostic features, required hepatic decompression procedures earlier, but had no impact on 5-year survival. Our results suggest that JAK2V617F testing should replace BM investigations as initial test for MPD in patients with SVT. Underlying MPD is associated with severe forms of BCS, but current therapy appears to offset deleterious effects of MPD on the medium-term outcome.


Assuntos
Janus Quinase 2/genética , Mutação , Receptores de Trombopoetina/genética , Circulação Esplâncnica/genética , Trombose Venosa/genética , Adulto , Exame de Medula Óssea , Síndrome de Budd-Chiari/complicações , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Transtornos Mieloproliferativos/complicações , Prognóstico , Estudos Retrospectivos , Trombose Venosa/mortalidade
8.
Blood ; 110(10): 3735-43, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17709604

RESUMO

The MPL (W515L and W515K) mutations have been detected in granulocytes of patients suffering from certain types of primitive myelofibrosis (PMF). It is still unknown whether this molecular event is also present in lymphoid cells and therefore potentially at the hematopoietic stem cell (HSC) level. Toward this goal, we conducted MPL genotyping of mature myeloid and lymphoid cells and of lymphoid/myeloid progenitors isolated from PMF patients carrying the W515 mutations. We detected both MPL mutations in granulocytes, monocytes, and platelets as well as natural killer (NK) cells but not in T cells. B/NK/myeloid and/or NK/myeloid CD34(+)CD38(-)-derived clones were found to carry the mutations. Long-term reconstitution of MPL W515 CD34(+) cells in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice was successful for as long as 12 weeks after transplantation, indicating that MPL W515 mutations were present in HSCs. Moreover, the 2 MPL mutations induced a spontaneous megakaryocytic growth in culture with an overall normal response to thrombopoietin (TPO). In contrast, erythroid progenitors remained EPO dependent. These results demonstrate that in PMF, the MPL W515L or K mutation induces a spontaneous megakaryocyte (MK) differentiation and occurs in a multipotent HSCs.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Mutação Puntual , Mielofibrose Primária/genética , Receptores de Trombopoetina/genética , Animais , Antígenos CD34/metabolismo , Sequência de Bases , Proliferação de Células , Células Cultivadas , Análise Mutacional de DNA , Frequência do Gene , Testes Genéticos/métodos , Genótipo , Humanos , Megacariócitos/citologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mielofibrose Primária/patologia , Receptores de Trombopoetina/metabolismo , Sensibilidade e Especificidade , Linfócitos T/metabolismo , Linfócitos T/patologia
9.
Blood ; 109(1): 71-7, 2007 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-16954506

RESUMO

The JAK2 V617F mutation has recently been described as an essential oncogenic event associated with polycythemia vera (PV), idiopathic myelofibrosis (IMF), and essential thrombocythemia. This mutation has been detected in all myeloid lineages but has not yet been detected in lymphoid cells. This raises the question whether this molecular event occurs in a true lymphomyeloid progenitor cell. In this work, we studied the presence of the mutation in peripheral blood cells and sorted B, T, and natural killer (NK) cells from PV and IMF. We detected the JAK2 V617F mutation in B and NK cells in approximately half the patients with IMF and a minority of those with PV. Moreover, in a few cases patients with IMF had mutated peripheral T cells. The mutation (homozygous or heterozygous) could be subsequently detected in B/NK/myeloid progenitors from PV and IMF, with a much higher frequency in clones derived from IMF. Using the fetal thymus organ culture (FTOC) assay, the mutation was also detected in all T-cell fractions derived from IMF and PV CD34+ cells. These results demonstrate that myeloproliferative disorders take their origin in a true myeloid/lymphoid progenitor cell but that their phenotype is related to a downstream selective proliferative advantage of the myeloid lineages.


Assuntos
Janus Quinase 2/genética , Linfócitos/enzimologia , Policitemia Vera/patologia , Mielofibrose Primária/patologia , Substituição de Aminoácidos , Animais , Antígenos CD34/análise , Linfócitos B/enzimologia , Diferenciação Celular , Divisão Celular , Linhagem Celular , Transformação Celular Neoplásica , Genótipo , Granulócitos/enzimologia , Células-Tronco Hematopoéticas/enzimologia , Células-Tronco Hematopoéticas/patologia , Humanos , Imunofenotipagem , Células Matadoras Naturais/enzimologia , Linfócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Células Mieloides/enzimologia , Transtornos Mieloproliferativos/patologia , Técnicas de Cultura de Órgãos , Fenótipo , Mutação Puntual , Policitemia Vera/enzimologia , Mielofibrose Primária/enzimologia , Seleção Genética , Linfócitos T/enzimologia , Timo/embriologia
10.
Cancer Res ; 65(8): 3281-9, 2005 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15833861

RESUMO

The release of transforming growth factor-beta1 (TGF-beta1) in the bone marrow microenvironment is one of the main mechanisms leading to myelofibrosis in murine models and probably in the human idiopathic myelofibrosis (IMF). The regulation of TGF-beta1 synthesis is poorly known but seems regulated by nuclear factor kappaB (NF-kappaB). We previously described the overexpression of an immunophilin, FK506 binding protein 51 (FKBP51), in IMF megakaryocytes. Gel shift and gene assays show that FKBP51's overexpression in a factor-dependent hematopoietic cell line, induces a sustained NF-kappaB activation after cytokine deprivation. This activation correlates with a low level of IkappaBalpha. A spontaneous activation of NF-kappaB was also detected in proliferating megakaryocytes and in circulating CD34(+) patient cells. In normal cells, NF-kappaB activation was only detected after cytokine treatment. The expression of an NF-kappaB superrepressor in FKBP51 overexpressing cells and in derived megakaryocytes from CD34(+) of IMF patients revealed that NF-kappaB activation was not involved in the resistance to apoptosis after cytokine deprivation of these cells but in TGF-beta1 secretion. These results highlight the importance of NF-kappaB's activation in the fibrosis development of this disease. They also suggest that FKBP51's overexpression in IMF cells could play an important role in the pathogenesis of this myeloproliferative disorder.


Assuntos
NF-kappa B/metabolismo , Mielofibrose Primária/metabolismo , Proteínas de Ligação a Tacrolimo/biossíntese , Fator de Crescimento Transformador beta/biossíntese , Antígenos CD34/biossíntese , Linhagem Celular Tumoral , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Mielofibrose Primária/sangue , Mielofibrose Primária/patologia , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta1
11.
Cancer Res ; 62(16): 4685-9, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12183426

RESUMO

Methionine depletion in the human cell line CCRF-CEM through the action of recombinant methioninase (rMETase), a methionine-cleaving enzyme, was previously demonstrated to produce a strong cytotoxic synergistic effect with fluorouracil (FUra) throughout a broad range of concentrations of FUra and rMETase, including subcytotoxic levels of rMETase. Potentiation was associated with a decrease in free thymidylate synthase from preexisting levels. To further investigate the action of rMETase on CCRF-CEM cells, in the present study we explored the effects of rMETase as a single agent on DNA methylation levels and DNA synthesis, which may be changed as a result of deprivation of methionine. Cells treated with rMETase under subcytotoxic conditions contained significantly lower levels of genomic methylated DNA than did control cells, as demonstrated by incorporation of the methyl radical of [methyl-(3)H]S-adenosylmethionine in DNA and by use of methylation-sensitive arbitrarily primed PCR. DNA hypomethylation produced by rMETase was of similar magnitude as that produced with the DNA methyltransferase inhibitor 5-azacytidine. Cells exposed to rMETase synthesized significantly more DNA than did untreated cells. Incorporation of [6-3H]thymidine and [6-3H]2'-deoxyuridine in these cells was augmented over that in control by mean factors of 1.78 and 2.36, respectively. Increased 3H nucleoside incorporation resulted in greater numbers of nuclear grains as demonstrated by autoradiography. The increase in DNA synthesis induced by rMETase is likely to result from enhancement of DNA repair because it was not accompanied by differences in cell cycle phase distribution or in total DNA content as determined by flow cytometry. We hypothesize that potentiation of FUra cytotoxicity by rMETase may result from increased inhibition of thymidylate synthase, together with DNA hypomethylation and enhanced DNA repair that could be involved in cell responses to drug-induced damage.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Liases de Carbono-Enxofre/farmacologia , Metilação de DNA/efeitos dos fármacos , DNA de Neoplasias/biossíntese , Leucemia de Células T/tratamento farmacológico , DNA de Neoplasias/genética , DNA de Neoplasias/metabolismo , Relação Dose-Resposta a Droga , Humanos , Cinética , Leucemia de Células T/genética , Leucemia de Células T/metabolismo , Proteínas Recombinantes/farmacologia , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA