Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 111(6): 2295-300, 2014 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-24469804

RESUMEN

The Janus kinase 2 (JAK2) V617F mutation is the primary pathogenic mutation in patients with Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs). Although thrombohemorrhagic incidents are the most common causes of morbidity and mortality in patients with MPNs, the events causing these clotting abnormalities remain unclear. To identify the cells responsible for the dysfunctional hemostasis, we used transgenic mice expressing JAK2V617F in specific lineages involved in thrombosis and hemostasis. When JAK2V617F was expressed in both hematopoietic and endothelial cells (ECs), the mice developed a significant MPN, characterized by thrombocytosis, neutrophilia, and splenomegaly. However, despite having significantly higher platelet counts than controls, these mice showed severely attenuated thrombosis following injury. Interestingly, platelet activation and aggregation in response to agonists was unaltered by JAK2V617F expression. Subsequent bone marrow transplants revealed the contribution of both endothelial and hematopoietic compartments to the attenuated thrombosis. Furthermore, we identified a potential mechanism for this phenotype through JAK2V617F-regulated inhibition of von Willebrand factor (VWF) function and/or secretion. JAK2V617F(+) mice display a condition similar to acquired von Willebrand syndrome, exhibiting significantly less high molecular weight VWF and reduced agglutination to ristocetin. These findings greatly advance our understanding of thrombohemorrhagic events in MPNs and highlight the critical role of ECs in the pathology of hematopoietic malignancies.


Asunto(s)
Trastornos de la Coagulación Sanguínea/enzimología , Endotelio Vascular/enzimología , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/complicaciones , Animales , Trastornos de la Coagulación Sanguínea/complicaciones , Plaquetas/patología , Ratones , Ratones Transgénicos , Receptor TIE-2/genética , Enfermedades de von Willebrand/genética
2.
Exp Hematol ; 38(5): 384-91, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20188141

RESUMEN

OBJECTIVE: To determine whether specific c-Mpl mutations might respond to thrombopoietin receptor agonists. MATERIALS AND METHODS: We created cell line models of type II c-Mpl mutations identified in congenital amegakaryocytic thrombocytopenia. We selected F104S c-Mpl for further study because it exhibited surface expression of the receptor. We measured proliferation of cell lines expressing wild-type or F104S c-Mpl in response to thrombopoietin receptor agonists targeting the extracellular (m-AMP4) or transmembrane (LGD-4665) domains of the receptor by 1-methyltetrazole-5-thiol assay. We measured thrombopoietin binding to the mutant receptor using an in vitro thrombopoietin uptake assay and identified F104 as a potentially critical residue for the interaction between the receptor and its ligand by aligning thrombopoietin and erythropoietin receptors from multiple species. RESULTS: Cells expressing F104S c-Mpl proliferated in response to LGD-4665, but not thrombopoietin or m-AMP4. Compared to thrombopoietin, LGD-4665 stimulates signaling with delayed kinetics in both wild-type and F104S c-Mpl-expressing cells. Although F104S c-Mpl is expressed on the cell surface in our BaF3 cell line model, the mutant receptor does not bind thrombopoietin. Comparison to the erythropoietin receptor suggests that F104 engages in hydrogen-bonding interactions that are critical for binding to thrombopoietin. CONCLUSIONS: These findings suggest that a small subset of patients with congenital amegakaryocytic thrombocytopenia might respond to treatment with thrombopoietin receptor agonists, but that responsiveness will depend on the type of mutation and agonist used. We postulate that F104 is critical for thrombopoietin binding. The kinetics of signaling in response to a transmembrane domain-binding agonist are delayed in comparison to thrombopoietin.


Asunto(s)
Péptidos/farmacología , Mutación Puntual , Receptores de Trombopoyetina/agonistas , Trombocitopenia/tratamiento farmacológico , Trombopoyesis/efectos de los fármacos , Animales , Línea Celular , Membrana Celular/metabolismo , Humanos , Enlace de Hidrógeno , Interleucina-3/farmacología , Linfocitos/metabolismo , Ratones , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Receptores de Trombopoyetina/química , Receptores de Trombopoyetina/genética , Proteínas Recombinantes de Fusión/agonistas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes/farmacología , Trombocitopenia/congénito , Trombocitopenia/genética , Trombocitopenia/patología , Trombopoyetina/metabolismo
3.
Exp Hematol ; 37(4): 495-503, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19302922

RESUMEN

OBJECTIVE: To genetically and functionally characterize mutations of c-Mpl that lead to thrombocytopenia in a child with congenital amegakaryocytic thrombocytopenia. MATERIALS AND METHODS: We identified two c-Mpl mutations in a child with clinical features of congenital amegakaryocytic thrombocytopenia, one a previously described mutation in the extracellular domain (R102P) and the other a novel mutation leading to truncation of the receptor after the box 1 homology domain (541Stop). Cell line models were created to examine the ability of the mutant receptors to signal in response to thrombopoietin and thrombopoietin-like agonists. RESULTS: Data from cell-line models indicate that c-Mpl R102P does not support significant signaling in response to thrombopoietin due to impaired trafficking of the mutant receptor to the cell surface. Alternative thrombopoietic agents do not circumvent this block to signaling, likely due to the inaccessibility of the receptor. In addition, previous data indicate that c-Mpl 541Stop does not support intracellular signaling due to the loss of critical intracellular domains. CONCLUSIONS: This case demonstrates two different mechanisms by which c-Mpl mutations can impair thrombopoietin signaling, and suggests that mutations in the extracellular domain will not be rescued by c-Mpl agonists if they interfere with normal receptor expression.


Asunto(s)
Heterocigoto , Receptores de Trombopoyetina/genética , Trombocitopenia/congénito , Trombocitopenia/genética , Secuencia de Bases , Línea Celular , Preescolar , Femenino , Glicosilación , Humanos , Megacariocitos/patología , Datos de Secuencia Molecular , Mutación , Unión Proteica , Receptores de Trombopoyetina/metabolismo , Transducción de Señal
4.
Cell Cycle ; 7(2): 257-66, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18256550

RESUMEN

Thrombopoietin (TPO), the primary regulator of megakaryocyte (MK) and platelet formation, modulates the activity of multiple signal transduction molecules, including those in the Jak/STAT, p42/p44 MAPK and phosphatidylinositol 3-kinase (PI3K)/Akt pathways. We previously demonstrated that PI3K and Akt are necessary for TPO-induced cell cycle progression of primary MK progenitors. However, the molecular events secondary to the activation of PI3K/Akt responsible for MK proliferation remain unclear. In this study we show that FOXO3a and its downstream target p27(Kip1) play an important role in TPO-induced proliferation of MK progenitors. We found that TPO down-modulates p27(Kip1) expression at both the mRNA and protein levels in primary MKs in a PI3K dependent fashion. UT-7/TPO, a megakaryocytic cell line, stably expressing constitutively active Akt or a dominant-negative form of FOXO3a failed to reduce p27(Kip1) expression after TPO stimulation, and fail to induce p27(Kip1) expression following TPO withdrawal. Induced expression of an active form of FOXO3a resulted in increased p27(Kip1) expression in this cell line. In addition, the number of MKs is significantly increased in bone marrow from Foxo3a-deficient mice. Taken together with the previous observation that p27(Kip1)-deficient mice also display increased numbers of MK progenitors, our findings indicate that the PI3K/Akt/FOXO3a/p27(Kip1) pathway contributes to normal TPO-induced MK proliferation.


Asunto(s)
Proliferación Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Factores de Transcripción Forkhead/metabolismo , Megacariocitos/citología , Trombopoyetina/metabolismo , Animales , Técnicas de Cultivo de Célula , Línea Celular , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Megacariocitos/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal
5.
Blood ; 111(2): 596-604, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17925492

RESUMEN

Focal adhesion kinase (FAK) plays a key role in mediating signaling downstream of integrins and growth factor receptors. In this study, we determined the roles of FAK in vivo by generating a megakaryocyte lineage-specific FAK-null mouse (Pf4-Cre/FAK-floxed). Megakaryocyte and platelet FAK expression was ablated in Pf4-Cre/FAK-floxed mice without affecting expression of the FAK homologue PYK2, although PYK2 phosphorylation was increased in FAK-/- megakaryocytes in response to fibrinogen. Megakaryopoiesis is greatly enhanced in Pf4-Cre/FAK-floxed mice, with significant increases in megakaryocytic progenitors (CFU-MK), mature megakaryocytes, megakaryocyte ploidy, and moderate increases in resting platelet number and platelet recovery following a thrombocytopenic stress. Thrombopoietin (Tpo)-mediated activation of Lyn kinase, a negative regulator of megakaryopoiesis, is severely attenuated in FAK-null megakaryocytes compared with wild-type controls. In contrast, Tpo-mediated activation of positive megakaryopoiesis regulators such as ERK1/2 and AKT is increased in FAK-null megakaryocytes, providing a plausible explanation for the observed increases in megakaryopoiesis in these mice. In Pf4-Cre/FAK-floxed mice, rebleeding times are significantly increased, and FAK-null platelets exhibit diminished spreading on immobilized fibrinogen. These studies establish clear roles for FAK in megakaryocyte growth and platelet function, setting the stage for manipulation of this component of the Tpo signaling apparatus for therapeutic benefit.


Asunto(s)
Plaquetas/enzimología , Quinasa 1 de Adhesión Focal/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Megacariocitos/enzimología , Trombopoyesis/fisiología , Trombopoyetina/metabolismo , Animales , Plaquetas/citología , Quinasa 1 de Adhesión Focal/genética , Megacariocitos/citología , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Ploidias , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Trombocitopenia/enzimología , Trombocitopenia/genética , Trombopoyetina/genética
6.
Cell Cycle ; 6(4): 455-60, 2007 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-17312391

RESUMEN

Megakaryocyte (MK) differentiation is marked by the development of progressive polyploidy, due to repeated incomplete cell cycles in which mitosis is aborted during anaphase, a process termed endomitosis. We have postulated that anaphase in endomitotic MKs diverges from diploid mitosis at a point distal to the assembly of the midzone, possibly involving impaired cleavage furrow progression. To define the extent of furrow initiation and ingression in endomitosis, we performed time-lapse imaging of MKs expressing yellow fluorescent protein (YFP)-tubulin and monitored shape change as they progressed through anaphase. We found that in early endomitotic cells that have a bipolar spindle, cleavage furrows form that can undergo significant ingression, but furrows regress to produce polyploid cells. Compared to cells that divide, cells that exhibit furrow regression have a slower rate of furrow ingression and do not furrow as deeply. More highly polyploid MKs undergoing additional endomitotic cycles also show measurable furrowing that is followed by regression, but the magnitude of the shape change is less than seen in the early MKs. This suggests that in the earliest endomitotic cycles when there is formation of a bipolar spindle, the failure of cytokinesis occurs late, following assembly and initial constriction of the actin/myosin ring, whereas in endomitotic MKs that are already polyploid there is secondary inhibition of furrow progression. This behavior of furrow ingression followed by regression may explain why midbody remnants are occasionally observed in polyploid MKs. This finding has important implications for the potential mechanisms for cytokinesis failure in endomitosis.


Asunto(s)
Megacariocitos/citología , Mitosis/fisiología , Huso Acromático/ultraestructura , Anafase/fisiología , Animales , Forma de la Célula , Células Cultivadas , Citocinesis/fisiología , Proteínas Luminiscentes/análisis , Megacariocitos/fisiología , Megacariocitos/ultraestructura , Ratones , Microscopía Fluorescente , Poliploidía , Proteínas Recombinantes de Fusión/análisis , Huso Acromático/fisiología , Tubulina (Proteína)/análisis
7.
Proc Natl Acad Sci U S A ; 103(15): 5965-70, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16585502

RESUMEN

Recombinant human erythropoietin (rhEPO) is receiving increasing attention as a potential therapy for prevention of injury and restoration of function in nonhematopoietic tissues. However, the minimum effective dose required to mimic and augment these normal paracrine functions of erythropoietin (EPO) in some organs (e.g., the brain) is higher than for treatment of anemia. Notably, a dose-dependent risk of adverse effects has been associated with rhEPO administration, especially in high-risk groups, including polycythemia-hyperviscosity syndrome, hypertension, and vascular thrombosis. Of note, several clinical trials employing relatively high dosages of rhEPO in oncology patients were recently halted after an increase in mortality and morbidity, primarily because of thrombotic events. We recently identified a heteromeric EPO receptor complex that mediates tissue protection and is distinct from the homodimeric receptor responsible for the support of erythropoiesis. Moreover, we developed receptor-selective ligands that provide tools to assess which receptor isoform mediates which biological consequence of rhEPO therapy. Here, we demonstrate that rhEPO administration in the rat increases systemic blood pressure, reduces regional renal blood flow, and increases platelet counts and procoagulant activities. In contrast, carbamylated rhEPO, a heteromeric receptor-specific ligand that is fully tissue protective, increases renal blood flow, promotes sodium excretion, reduces injury-induced elevation in procoagulant activity, and does not effect platelet production. These preclinical findings suggest that nonerythropoietic tissue-protective ligands, which appear to elicit fewer adverse effects, may be especially useful in clinical settings for tissue protection.


Asunto(s)
Coagulación Sanguínea/efectos de los fármacos , Coagulantes/farmacología , Eritropoyetina/análogos & derivados , Eritropoyetina/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/fisiología , Línea Celular , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Eritropoyetina/uso terapéutico , Hematopoyesis/efectos de los fármacos , Hemodinámica/efectos de los fármacos , Humanos , Ratones , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología , Células Madre/efectos de los fármacos , Células Madre/fisiología , Venas Umbilicales
8.
Exp Hematol ; 34(1): 82-6, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413394

RESUMEN

OBJECTIVE: Blood levels of thrombopoietin (TPO) are regulated in part by cellular degradation following its binding to the cell surface receptor c-mpl. Previous reports have demonstrated that in addition to hematopoietic cells, c-mpl is expressed on and functions in several types of endothelial cells (ECs). We hypothesized that the c-mpl expressed on ECs would contribute to the regulation of circulating TPO levels. METHODS: To test this hypothesis we transplanted c-mpl-null and wild-type (WT) control mice with WT marrow stem cells, resulting in two groups of posttransplant chimeric animals, one expressing c-mpl on megakaryocytes and platelets only and one in which the receptor is expressed on both hematopoietic and ECs. Should EC c-mpl take up TPO and degrade it, we predicted that c-mpl-null mice reconstituted with WT cells would display increased TPO levels and an increased steady state platelet count compared to the WT recipients. RESULTS: Contrary to our prediction, for up to 6 months posttransplantation both platelet counts and TPO levels in both groups of transplanted mice were virtually identical. CONCLUSIONS: Our results indicate that the EC c-mpl receptor does not contribute significantly to the regulation of TPO levels or to steady-state platelet counts. These results also imply that patients with congenital amegakaryocytic thrombocytopenia, lacking the c-mpl receptor, who have successfully been engrafted with normal hematopoietic stem cells should have normal (not elevated) TPO levels and that gene replacement strategies designed to restore c-mpl in these patients do not need to target ECs to establish the normal regulation of TPO.


Asunto(s)
Células Endoteliales/metabolismo , Proteínas Oncogénicas/genética , Receptores de Citocinas/genética , Trombopoyetina/sangre , Trombopoyetina/metabolismo , Animales , Línea Celular , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/metabolismo , Recuento de Plaquetas , Receptores de Citocinas/metabolismo , Receptores de Trombopoyetina , Trasplante de Células Madre
9.
Cell Signal ; 18(8): 1212-8, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16380230

RESUMEN

Thrombopoietin (TPO) and its receptor (c-Mpl) are the major regulators of megakaryocyte and platelet production and serve a critical and non-redundant role in hematopoietic stem cell (HSC) biology. TPO signals through the Jak-STAT, Ras-Raf-MAPK, and PI3K pathways, and promotes survival, proliferation, and polyploidization in megakaryocytes. The proto-oncogene c-myc also plays an important role in many of these same processes. In this work we studied the regulated expression of c-myc in megakaryocytic cell lines and primary cells by quantitative real-time RT-PCR. We found that TPO induced expression of c-myc in 1 h in both hematopoietic cell lines (UT-7 and BaF3/Mpl) and mature murine megakaryocytes. The TPO-induced expression of c-myc was blocked by a phosphatidylinositol 3-kinase (PI3K) inhibitor, suggesting that TPO stimulated c-myc expression through a PI3K-dependent pathway. Of interest, our study showed that overexpression of active Akt did not rescue the effect of PI3K blockade on c-myc expression, rather, enhanced it. In addition, inhibitors of protein kinase C (PKC)zeta and the target of rapamycin (mTOR) also failed to affect c-myc mRNA expression, while c-myc mRNA expression was reduced by inhibition of the mitogen activated protein kinase (MAPK) pathway. Therefore, we conclude that TPO stimulates c-myc expression in primary megakaryocytes through a PI3K- and MAPK-dependent pathway that is not mediated by Akt, PKCzeta or mTOR.


Asunto(s)
Regulación de la Expresión Génica/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Trombopoyetina/farmacología , Animales , Línea Celular , Cricetinae , Femenino , Humanos , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteína Quinasa C/antagonistas & inhibidores , Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , ARN Mensajero/genética , ARN Mensajero/metabolismo , Serina-Treonina Quinasas TOR
10.
Exp Hematol ; 33(1): 94-9, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15661402

RESUMEN

OBJECTIVE: Studies in numerous adherent cell systems have indicated that engagement of integrins is required for cell survival and proliferation. Although not classically thought of as an adherent cell type, megakaryocytes in the marrow develop in juxtaposition to endothelial cells which display a number of integrin counter-receptors. Moreover, a number of other hematopoietic cell types, including stem cells and erythroid progenitors, have been shown to engage and be affected by integrin ligands. METHODS: The role of beta1 integrins in thrombopoietin-mediated megakaryopoiesis was studied using both gain-of-function and loss-of-function strategies. RESULTS: We found that pan-blockade of integrins with a relatively nonspecific disintegrin blocked TPO-induced MK growth, but that an alpha5beta1 disintegrin, and a function-blocking monoclonal antibody, failed to affect megakaryopoiesis in vitro. In contrast, a neutralizing alpha4beta1 monoclonal antibody blocked TPO-induced MK growth, and an integrin alpha4beta1 ligand, the H296 fragment of fibronectin, enhanced MK growth at all concentrations of TPO. CONCLUSIONS: These findings have important implications for thrombopoiesis in general, and potentially for the enhanced platelet production found in states of systemic inflammation and following the use of therapeutic strategies designed to block alpha4beta1 integrin engagement in states of chronic inflammation and autoimmunity.


Asunto(s)
Integrina alfa4beta1/fisiología , Trombopoyesis , Trombopoyetina/fisiología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Células de la Médula Ósea/citología , Proliferación Celular/efectos de los fármacos , Desintegrinas/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/citología , Fibronectinas/metabolismo , Fibronectinas/farmacología , Integrina alfa4beta1/antagonistas & inhibidores , Integrina alfa4beta1/metabolismo , Megacariocitos/citología , Ratones , Trombopoyesis/efectos de los fármacos , Trombopoyetina/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...