Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
1.
Haemophilia ; 23(5): 784-792, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28662545

RESUMEN

BACKGROUND: RUNX1 haplodeficiency is associated with thrombocytopenia, platelet dysfunction and a predisposition to acute leukaemia. Platelets possess three distinct types of granules and secretory processes involving dense granules (DG), α-granules and vesicles or lysosomes containing acid hydrolases (AH). Dense granules and granule deficiencies have been reported in patients with RUNX1 mutations. Little is known regarding the secretion from AH-containing vesicles. METHODS AND RESULTS: We studied two related patients with a RUNX1 mutation, easy bruising, and mild thrombocytopenia. Platelet aggregation and 14 C serotonin in platelet-rich plasma (PRP) were impaired in response to ADP, epinephrine, collagen and arachidonic acid. Contents of DG (ATP, ADP), α-granules (ß-thromboglobulin) and AH-containing vesicles (ß-glucuronidase, ß-hexosaminidase, α-mannosidase) were normal or minimally decreased. Dense granules secretion on stimulation of gel-filtered platelets with thrombin and divalent ionophore A23187 (4-12 µmol L-1 ) were diminished. ß-thromboglobulin and AH secretion was impaired in response to thrombin or A23187. We studied thromboxane-related pathways. The incorporation of 14 C -arachidonic acid into phospholipids and subsequent arachidonic acid release on thrombin activation was normal. Platelet thromboxane A2 production in whole blood serum and on thrombin stimulation of PRP was normal, suggesting that the defective secretion was not due to impaired thromboxane production. CONCLUSIONS: These studies provide the first evidence in patients with a RUNX1 mutation for a defect in AH (lysosomal) secretion, and for a global defect in secretion involving all three types of platelet granules that is unrelated to a granule content deficiency. They highlight the pleiotropic effects and multiple platelet defects associated with RUNX1 mutations.


Asunto(s)
Trastornos de las Plaquetas Sanguíneas/genética , Trastornos de las Plaquetas Sanguíneas/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Haploinsuficiencia , Hidrolasas/metabolismo , Adulto , Ácido Araquidónico/metabolismo , Trastornos de las Plaquetas Sanguíneas/sangre , Plaquetas/metabolismo , Estudios de Casos y Controles , Preescolar , Femenino , Humanos , Masculino , Mutación , Fosfolípidos/biosíntesis , Recuento de Plaquetas , Vesículas Secretoras/metabolismo , Serotonina/metabolismo , Tromboxanos/biosíntesis
2.
J Thromb Haemost ; 13(10): 1888-99, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26256688

RESUMEN

BACKGROUND: Megakaryocytes express and store platelet factor 4 (PF4) in alpha granules. In vivo, PF4 is a clinically relevant, negative regulator of megakaryopoiesis and hematopoietic stem cell replication. These findings would suggest a regulated source of free intramedullary PF4. OBJECTIVES: Define the source of free intramedullary PF4 and its intramedullary life cycle. METHODS: We interrogated both murine and human bone marrow-derived cells during megakaryopoiesis in vitro by using confocal microscopy and enzyme-linked immunosorbent assay. With immunohistochemistry, we examined in vivo free PF4 in murine bone marrow before and after radiation injury and in the setting of megakaryocytopenia and thrombocytopenia. RESULTS: Exogenously added human PF4 is internalized by murine megakaryocytes. Human megakaryocytes similarly take up murine PF4 but not the related chemokine, platelet basic protein. Confocal microscopy shows that internalized PF4 colocalizes with endogenous PF4 in alpha granules and is available for release on thrombin stimulation. Immunohistochemistry shows free PF4 in the marrow, but not another alphagranule protein, von Willebrand factor. Free PF4 increases with radiation injury and decreases with megakaryocytopenia. Consistent with the known role of low-density lipoprotein receptor-related protein 1 in the negative paracrine effect of PF4 on megakaryopoiesis, PF4 internalization is at least partially low-density lipoprotein receptor-related protein 1 dependent. CONCLUSIONS: PF4 has a complex intramedullary life cycle with important implications in megakaryopoiesis and hematopoietic stem cell replication not seen with other tested alpha granule proteins.


Asunto(s)
Gránulos Citoplasmáticos/metabolismo , Megacariocitos/metabolismo , Factor Plaquetario 4/metabolismo , Trombocitopenia/metabolismo , Trombopoyesis , Animales , Transporte Biológico , Células Cultivadas , Gránulos Citoplasmáticos/efectos de la radiación , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Inmunohistoquímica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Megacariocitos/efectos de la radiación , Ratones Noqueados , Microscopía Confocal , Factor Plaquetario 4/deficiencia , Factor Plaquetario 4/genética , Interferencia de ARN , Receptores de LDL/genética , Receptores de LDL/metabolismo , Trombocitopenia/sangre , Trombocitopenia/genética , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
3.
J Thromb Haemost ; 13 Suppl 1: S143-50, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26149015

RESUMEN

We have proposed that modified platelets could potentially be used to correct intrinsic platelet defects as well as for targeted delivery of therapeutic molecules to sights of vascular injury. Ectopic expression of proteins within α-granules prior to platelet activation has been achieved for several proteins, including urokinase, factor (F) VIII, and partially for FIX. Potential uses of platelet-directed therapeutics will be discussed, focusing on targeted delivery of urokinase as a thromboprophylactic agent and FVIII for the treatment of hemophilia A patients with intractable inhibitors. This presentation will discuss new strategies that may be useful in the care of patients with vascular injury as well as remaining challenges and limitations of these approaches.


Asunto(s)
Trastornos de la Coagulación Sanguínea/terapia , Trastornos de las Plaquetas Sanguíneas/terapia , Plaquetas/metabolismo , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Transfusión de Plaquetas , Animales , Trastornos de la Coagulación Sanguínea/sangre , Trastornos de la Coagulación Sanguínea/genética , Factores de Coagulación Sanguínea/biosíntesis , Factores de Coagulación Sanguínea/genética , Trastornos de las Plaquetas Sanguíneas/sangre , Trastornos de las Plaquetas Sanguíneas/genética , Deficiencia del Factor V/sangre , Deficiencia del Factor V/genética , Deficiencia del Factor V/terapia , Predisposición Genética a la Enfermedad , Hemofilia A/sangre , Hemofilia A/genética , Hemofilia A/terapia , Humanos , Resultado del Tratamiento , Activador de Plasminógeno de Tipo Uroquinasa/biosíntesis , Activador de Plasminógeno de Tipo Uroquinasa/sangre , Activador de Plasminógeno de Tipo Uroquinasa/genética
4.
J Thromb Haemost ; 13(8): 1416-27, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25960020

RESUMEN

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is an iatrogenic complication of heparin therapy caused by antibodies to a self-antigen, platelet factor (4) and heparin. The reasons why antibodies form to PF4/heparin, but not to PF4 bound to other cellular glycosaminoglycans are poorly understood. OBJECTIVE: To investigate differences in cellular responses to cell-bound PF4 and PF4/heparin complexes, we studied the internalization of each by peripheral blood-derived monocytes, dendritic cells and neutrophils. METHODS AND RESULTS: Using unlabeled and fluorescently-labeled antigen and/or labeled monoclonal antibody to PF4/heparin complexes (KKO), we show that PF4/heparin complexes are taken up by monocytes in a heparin-dependent manner and are internalized by human monocytes and dendritic cells, but not by neutrophils. Complexes of PF4/low-molecular-weight heparin and complexes composed of heparin and murine PF4, protamine or lysozyme are internalized similarly, suggesting a common endocytic pathway. Uptake of complexes is mediated by macropinocytosis, as shown by inhibition using cytochalasin D and amiloride. Internalized complexes are transported intact to late endosomes, as indicated by co-staining of vesicles with KKO and lysosomal associated membrane protein-2 (LAMP-2). Lastly, we show that cellular uptake is accompanied by expression of MHCII and CD83 co-stimulatory molecules. CONCLUSIONS: Taken together, these studies establish a distinct role for heparin in enhancing antigen uptake and activation of the initial steps in the cellular immune response to PF4-containing complexes.


Asunto(s)
Anticoagulantes/toxicidad , Heparina/toxicidad , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Fagocitosis/efectos de los fármacos , Pinocitosis/efectos de los fármacos , Factor Plaquetario 4/metabolismo , Anticoagulantes/inmunología , Anticoagulantes/metabolismo , Antígenos CD/metabolismo , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Endosomas/efectos de los fármacos , Endosomas/metabolismo , Heparina/inmunología , Heparina/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inmunoglobulinas/metabolismo , Proteína 2 de la Membrana Asociada a los Lisosomas/metabolismo , Lisosomas/efectos de los fármacos , Lisosomas/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Glicoproteínas de Membrana/metabolismo , Monocitos/inmunología , Monocitos/metabolismo , Factor Plaquetario 4/inmunología , Unión Proteica , Factores de Tiempo , Antígeno CD83
5.
J Thromb Haemost ; 12(12): 2102-12, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25287191

RESUMEN

BACKGROUND: Ectopically expressed B-domainless factor VIII in megakaryocytes is stored in α-granules, is effective in a number of murine hemostatic models, and is protected from circulating inhibitors. However, this platelet (p) FVIII has different temporal-spatial availability from plasma FVIII, with limited efficacy in other murine hemostatic models. OBJECTIVES AND METHODS: We sought to improve pFVIII hemostatic efficacy by expressing canine (c) FVIII, which has higher stability and activity than human (h) FVIII in FVIII(null) mice. RESULTS AND CONCLUSIONS: We found that pcFVIII was more effective than phFVIII at restoring hemostasis, but peak pcFVIII antigen levels were lower and were associated with greater megakaryocyte apoptosis than phFVIII. These new insights suggest that pFVIII gene therapy strategies should focus on enhancing activity rather than levels. We previously showed that modification of the PACE/furin cleavage site in hFVIII resulted in secretion of hFVIII primarily as a single-chain molecule with increased biological activity. In megakaryocytes, this variant was expressed at the same level as phFVIII with a lentiviral bone marrow transplant approach to reconstitute FVIII(null) mice, but was more effective, resulting in near-normal hemostasis in the cremaster laser injury model. These studies may have implications for pFVIII gene therapy in hemophilia A.


Asunto(s)
Apoptosis , Plaquetas/citología , Factor VIII/química , Factor VIII/genética , Terapia Genética/métodos , Megacariocitos/citología , Animales , Arterias Carótidas/patología , Línea Celular , Cricetinae , Perros , Hemofilia A/genética , Hemostasis , Humanos , Lentivirus/genética , Ratones , Ratones Transgénicos
6.
J Thromb Haemost ; 12(11): 1890-7, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25156521

RESUMEN

BACKGROUND: ERp57 is required for platelet function; however, whether ERp57 contributes to fibrin generation is unknown. METHODS AND RESULTS: Using an inhibitory anti-ERp57 antibody (mAb1), Pf4-Cre/ERp57(fl/fl) mice, Tie2-Cre/ERp57(fl/fl) mice, and mutants of ERp57, we analyzed the function of ERp57 in laser-induced thrombosis. Fibrin deposition was decreased in Pf4-Cre/ERp57(fl/fl) mice, consistent with a role for platelet ERp57 in fibrin generation. Fibrin deposition was further decreased with infusion of mAb1 and in Tie2-Cre/ERp57(fl/fl) mice, consistent with endothelial cells also contributing to fibrin deposition. Infusion of eptibifatide inhibited platelet and fibrin deposition, confirming a role for platelets in fibrin deposition. Infusion of recombinant ERp57 corrected the defect in fibrin deposition but not platelet accumulation, suggesting a direct effect of ERp57 on coagulation. mAb1 inhibited thrombin generation in vitro, consistent with a requirement for ERp57 in coagulation. Platelet accumulation was decreased to similar extents in Pf4-Cre/ERp57(fl/fl) mice, Tie2-Cre/ERp57(fl/fl) mice and normal mice infused with mAb1. Infusion of completely inactivated ERp57 or ERp57 with a non-functional second active site inhibited fibrin deposition and platelet accumulation, indicating that the isomerase activity of the second active site is required for these processes. CONCLUSION: ERp57 regulates thrombosis via multiple targets.


Asunto(s)
Coagulación Sanguínea , Fibrina/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Trombosis/enzimología , Animales , Anticuerpos Monoclonales/farmacología , Plaquetas/enzimología , Modelos Animales de Enfermedad , Células Endoteliales/enzimología , Fibrinolíticos/farmacología , Terapia por Láser , Ratones Noqueados , Agregación Plaquetaria , Inhibidores de Agregación Plaquetaria/farmacología , Proteína Disulfuro Isomerasas/deficiencia , Proteína Disulfuro Isomerasas/genética , Proteína Disulfuro Isomerasas/inmunología , Transducción de Señal , Trombosis/sangre , Trombosis/etiología , Trombosis/genética , Trombosis/prevención & control , Factores de Tiempo
9.
J Thromb Haemost ; 9(10): 2077-86, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21848641

RESUMEN

BACKGROUND: Stromal interaction molecule 1 (STIM1) was recently identified as a critical component of store-operated calcium entry (SOCE) in platelets. We previously reported the Ca(2+) -sensing guanine nucleotide exchange factor CalDAG-GEFI as a critical molecule in Ca(2+) signaling in platelets. OBJECTIVE: To evaluate the contribution of STIM1/SOCE to Ca(2+) -dependent platelet activation and thrombosis, we here compared the activation responses of platelets lacking STIM1 and platelets lacking CalDAG-GEFI. METHODS: The murine Stim1 gene was conditionally deleted in the megakaryocyte/platelet lineage. CalDAG-GEFI(-/-) and Stim1(fl/fl) PF4-Cre mice, along with littermate control mice, were used for in vitro and in vivo experiments under flow as well as static conditions. RESULTS: Integrin α(IIb) ß(3) -mediated aggregation was markedly impaired in CalDAG-GEFI-deficient but not STIM1-deficient platelets, under both static and flow conditions. In contrast, deficiency in either STIM1 or CalDAG-GEFI significantly impaired the ability of platelets to express phosphatidylserine on the cell surface. When subjected to a laser injury thrombosis model, mice lacking STIM1 in platelets were characterized by the formation of unstable platelet-rich thrombi and delayed and reduced fibrin generation in injured arterioles. In CalDAG-GEFI(-/-) mice, fibrin generation was also delayed and reduced, but platelet accumulation was almost abolished. CONCLUSIONS: Our studies suggest that: (i) STIM1/SOCE is critical for the procoagulant activity but not the proadhesive function of platelets; and (ii) at the site of vascular injury, STIM1 and CalDAG-GEFI are critical for the first wave of thrombin generation mediated by procoagulant platelets.


Asunto(s)
Calcio/metabolismo , Factores de Intercambio de Guanina Nucleótido/fisiología , Glicoproteínas de Membrana/fisiología , Activación Plaquetaria , Trombosis/fisiopatología , Animales , Western Blotting , Canales de Calcio , Masculino , Ratones , Molécula de Interacción Estromal 1
11.
J Thromb Haemost ; 7 Suppl 1: 249-52, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19630810

RESUMEN

Heparin-induced thrombocytopenia (HIT) is an iatrogenic disorder that occurs in a small subset of patients receiving heparin. Twenty-five per cent (or higher) of affected patients develop limb or life-threatening thrombosis. The effectiveness of therapy is incomplete and may be complicated by bleeding. HIT is caused by antibodies that recognize the platelet chemokine, Platelet Factor 4 (PF4), complexed to heparin or to cellular glycosaminoglycans (GAGs). However, antibodies with the same apparent specificity are found in many more patients without clinical disease and the reason why so few develop HIT is uncertain. We propose that HIT antibodies recognize cell surface PF4/GAG complexes on intravascular cells, including platelets and monocytes that are dynamic and mutable. Heparin removes cell surface-bound PF4 in most individuals, but removal is incomplete in those with high pre-exposure surface-bound PF4 levels. Such individuals retain critically localized cellular antigenic complexes at the time antibodies develop and are at risk to develop HIT. This article reviews the scientific basis for this model and its clinical implications.


Asunto(s)
Plaquetas/inmunología , Heparina/efectos adversos , Monocitos/inmunología , Trombocitopenia/etiología , Anticuerpos , Plaquetas/química , Glicosaminoglicanos/metabolismo , Humanos , Monocitos/química , Factor Plaquetario 4/metabolismo , Trombocitopenia/inducido químicamente
12.
J Thromb Haemost ; 6(7): 1160-6, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18433455

RESUMEN

BACKGROUND: Gene therapy strategies directed at expressing factor (F)VIII in megakaryocytes has potential advantages in the treatment of hemophilia A. Among these is that platelet (p) FVIII may be effective in the presence of circulating anti-FVIII inhibitors. OBJECTIVE: We examined in a murine transgenic model whether pFVIII could correct the coagulation defect in FVIII(null) mouse in the presence of circulating inhibitors. METHODS: FVIII(null) mice that were transgenic for pFVIII (pFVIII/FVIII(null)) were compared with FVIII(null) mice receiving infused FVIII in a FeCl(3) carotid injury model in the presence of anti-FVIII inhibitors. RESULTS: After injury, pFVIII/FVIII(null) mice were significantly more resistant to circulating inhibitors than after plasma FVIII correction in both an acute and chronic models of inhibitor exposure even although in the chronic model, significant amounts of inhibitor were stored within the platelets. Furthermore, bleeding in the pFVIII mice in the presence of inhibitors was not as a result of the development of thrombocytopenia. CONCLUSION: In FVIII(null) mice, pFVIII provides improved, but limited, protection in the presence of inhibitors of approximately 6-fold greater Bethesda Units per mL relative to infused FVIII. Our findings differ from a recent report using a tail-clip exsanguination assay on the degree of efficacy of pFVIII in the presence of inhibitors. We propose that this difference in outcome is as a result of the sensitivity of the tail-vein exsanguination model to low levels of pFVIII.


Asunto(s)
Anticuerpos/efectos de los fármacos , Plaquetas/metabolismo , Factor VIII/administración & dosificación , Factor VIII/inmunología , Terapia Genética/métodos , Tolerancia Inmunológica/inmunología , Animales , Anticuerpos/sangre , Enfermedades de las Arterias Carótidas/terapia , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Ratones Transgénicos
13.
J Thromb Haemost ; 3(12): 2730-7, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16359510

RESUMEN

The role of factor V Leiden (FVL) as a modifier of the severe hemophilia phenotype is still unclear. We used mice with hemophilia A or B crossed with FVL to elucidate in vivo parameters of hemostasis. Real-time thrombus formation in the microcirculation was monitored by deposition of labeled platelets upon laser-induced endothelial injury using widefield microscopy in living animals. No thrombi formed in hemophilic A or B mice following vascular injuries. However, hemophilic mice, either heterozygous or homozygous for FVL, formed clots at all injured sites. Injection of purified activated FV into hemophilic A or B mice could mimic the in vivo effect of FVL. In contrast to these responses to a laser injury in a microvascular bed, FVL did not provide sustained hemostasis following damage of large vessels in a ferric chloride carotid artery injury model, despite of the improvement of clotting times and high circulating thrombin levels. Together these data provide evidence that FVL has the ability to improve the hemophilia A or B phenotype, but this effect is principally evident at the microcirculation level following a particular vascular injury. Our observations may partly explain the heterogeneous clinical evidence of the beneficial role of FVL in hemophilia.


Asunto(s)
Factor V/fisiología , Hemofilia A/sangre , Hemostasis , Animales , Coagulación Sanguínea/genética , Traumatismos de las Arterias Carótidas/sangre , Modelos Animales de Enfermedad , Hemofilia A/genética , Hemofilia B , Hemostasis/genética , Ratones , Ratones Endogámicos , Microcirculación , Microscopía por Video , Músculo Esquelético/irrigación sanguínea
14.
Blood ; 98(8): 2442-7, 2001 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-11588041

RESUMEN

Heparin-induced thrombocytopenia/thrombosis (HIT/HITT) is a severe, life-threatening complication that occurs in 1% to 3% of patients exposed to heparin. Interactions between heparin, human platelet factor 4 (hPF4), antibodies to the hPF4/heparin complex, and the platelet Fc receptor (FcR) for immunoglobulin G, FcgammaRIIA, are the proposed primary determinants of the disease on the basis of in vitro studies. The goal of this study was to create a mouse model that recapitulates the disease process in humans in order to understand the factors that predispose some patients to develop thrombocytopenia and thrombosis and to investigate new therapeutic approaches. Mice that express both human platelet FcgammaRIIA and hPF4 were generated. The FcgammaRIIA/hPF4 mice and controls, transgenic for either FcgammaRIIA or hPF4, were injected with KKO, a mouse monoclonal antibody specific for hPF4/heparin complexes, and then received heparin (20 U/d). Nadir platelet counts for KKO/heparin-treated FcgammaRIIA/hPF4 mice were 80% below baseline values, significantly different (P <.001) from similarly treated controls. FcgammaRIIA/hPF4 mice injected with KKO and 50 U/d heparin developed shock and showed fibrin-rich thrombi in multiple organs, including thrombosis in the pulmonary vasculature. This is the first mouse model of HIT to recapitulate the salient features of the human disease and demonstrates that FcgammaRIIA and hPF4 are both necessary and sufficient to replicate HIT/HITT in an animal model. This model should facilitate the identification of factors that modulate disease expression and the testing of novel therapeutic interventions.


Asunto(s)
Antígenos CD/metabolismo , Heparina/toxicidad , Factor Plaquetario 4/metabolismo , Receptores de IgG/metabolismo , Trombocitopenia/inducido químicamente , Trombosis/inducido químicamente , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Transgénicos , Activación Plaquetaria , Recuento de Plaquetas , Trombocitopenia/genética , Trombocitopenia/patología , Trombosis/genética , Trombosis/patología
15.
Thromb Haemost ; 86(3): 894-901, 2001 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11583324

RESUMEN

The peptide LSARLAF causes alphaIIbeta3-dependent platelet activation exemplified by secretion, aggregation, spreading and adhesion on fibrinogen, and tyrosine phosphorylation. alphaIIIbeta3-dependent outside-in signal transduction induced by LSARLAF was investigated in variant thrombasthenic platelets which lack most of the cytoplasmic domain of the integrin beta3 subunit (alphaIIbbeta3 delta724). These studies revealed that only certain aspects of this alphaIIbbeta3-dependent outside-in signaling were affected by the beta3 truncation. Specifically, alphaIIbbeta3 delta724 supported LSARLAF-induced platelet aggregation, agglutination and secretion, but failed to trigger cytoskeletal reorganization and platelet spreading on fibrinogen, despite the fact that PMA-induced non alphaIIbbeta3 mediated signaling caused spreading of these platelets on fibrinogen. Thus, distinct domains of alphaIIbbeta3 are required to support different aspects of LSARLAF-induced platelet activation. Furthermore, these studies suggest that not all alphaIIbbeta3-dependent platelet responses require an intact beta3 cytoplasmic tail.


Asunto(s)
Antígenos CD/genética , Codón sin Sentido , Mutación del Sistema de Lectura , Oligopéptidos/farmacología , Activación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/genética , Transducción de Señal/efectos de los fármacos , Trombastenia/genética , Adenosina Difosfato/farmacología , Adolescente , Alelos , Antígenos CD/química , Antígenos CD/fisiología , Epinefrina/farmacología , Trastornos Hemorrágicos/sangre , Trastornos Hemorrágicos/genética , Humanos , Integrina beta3 , Sustancias Macromoleculares , Masculino , Fosforilación/efectos de los fármacos , Activación Plaquetaria/fisiología , Agregación Plaquetaria/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/fisiología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/fisiología , Eliminación de Secuencia , Transducción de Señal/fisiología , Relación Estructura-Actividad , Acetato de Tetradecanoilforbol/farmacología , Trombastenia/sangre , Trombina/farmacología
16.
Blood ; 98(3): 610-7, 2001 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-11468158

RESUMEN

The genes for the related human (h) chemokines, PBP (platelet basic protein) and PF4 (platelet factor 4), are within 5.3 kilobases (kb) of each other and form a megakaryocyte-specific gene locus. The hypothesis was considered that the PBP and PF4 genes share a common distal regulatory region(s) that leads to their high-level megakaryocyte-specific expression in vivo. This study examined PBP and PF4 expression in transgenic mice using 4 distinct human PBP/PF4 gene locus constructs. These studies showed that within the region studied there was sufficient information to regulate tissue-specific expression of both hPBP and hPF4. Indeed this region contained sufficient DNA information to lead to expression levels of PBP and PF4 comparable to the homologous mouse genes in a position-independent, copy number-dependent fashion. These studies also indicated that the DNA domains that led to this expression were distinct for the 2 genes; hPBP expression is regulated by a region that is 1.5 to 4.4 kb upstream of that gene. Expression of hPF4 is regulated by a region that is either intergenic between the 2 genes or immediately downstream of the hPF4 gene. Comparison of the available human and mouse sequences shows conserved flanking region domains containing potential megakaryocyte-related transcriptional factor DNA-binding sites. Further analysis of these regulatory regions may identify enhancer domains involved in megakaryopoiesis that may be useful in the selective expression of other genes in megakaryocytes and platelets as a strategy for regulating hemostasis, thrombosis, and inflammation. (Blood. 2001;98:610-617)


Asunto(s)
Quimiocinas/genética , Genes Reguladores/genética , Megacariocitos/metabolismo , Factor Plaquetario 4/genética , Animales , Secuencia de Bases , Sitios de Unión , Quimiocinas/química , Secuencia Conservada , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Factor Plaquetario 4/metabolismo , Estructura Terciaria de Proteína , Bazo/química , Bazo/citología , Factores de Transcripción , beta-Tromboglobulina
17.
Ann Thorac Surg ; 71(6): 1920-4, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11426769

RESUMEN

BACKGROUND: Studies have demonstrated a high incidence of antibodies to heparin/platelet factor 4 complexes, the antigen in heparin-induced thrombocytopenia, in patients after cardiopulmonary bypass surgery. In many hospitals, beef lung heparin has been used historically for cardiopulmonary bypass, and there has been reluctance to change to porcine heparin despite concerns of an increased incidence of heparin-induced thrombocytopenia in patients receiving bovine heparin. METHODS: A prospective randomized trial comparing bovine and porcine heparin in cardiopulmonary bypass surgery was conducted. Presurgery and postsurgery heparin antibody formation was studied using the serotonin release assay and a heparin/platelet factor 4 enzyme-linked immunosorbent assay. RESULTS: Data available on 98 patients, randomized to receive either bovine or porcine heparin, revealed no significant difference in patient positivity by serotonin release assay (12% in both groups) or by the heparin/platelet factor 4 enzyme-linked immunosorbent assay (29% with porcine and 35% with bovine heparin) postoperatively. There were no significant differences between preoperative and postoperative platelet counts or thromboembolic complications. CONCLUSIONS: Our study does not support the belief that bovine heparin is more likely than porcine heparin to induce the development of antibodies to heparin/platelet factor 4.


Asunto(s)
Puente Cardiopulmonar , Heparina/efectos adversos , Trombocitopenia/inducido químicamente , Anciano , Animales , Anticuerpos/sangre , Bovinos , Femenino , Heparina/administración & dosificación , Heparina/inmunología , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo , Porcinos , Trombocitopenia/sangre
18.
J Biol Chem ; 276(17): 13975-81, 2001 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-11278919

RESUMEN

To determine the molecular basis for the insensitivity of rat alpha(IIb)beta(3) to inhibition by RGD-containing peptides, hybrids of human and rat alpha(IIb)beta(3) and chimeras of alpha(IIb)beta(3) in which alpha(IIb) was composed of portions of human and rat alpha(IIb) were expressed in Chinese hamster ovary cells and B lymphocytes, and the ability of the tetrapeptide RGDS to inhibit fibrinogen binding to the various forms of alpha(IIb)beta(3) was measured. These measurements indicated that sequences regulating the sensitivity of alpha(IIb)beta(3) to RGDS are located in the seven amino-terminal repeats of alpha(IIb). Moreover, replacing the first three or four (but not the first two) repeats of rat alpha(IIb) with the corresponding human sequences enhanced sensitivity to RGDS, whereas replacing the first two or three repeats of human alpha(IIb) with the corresponding rat sequences had little or no effect. Nevertheless, RGDS bound to Chinese hamster ovary cells expressing alpha(IIb)beta(3) regardless whether the alpha(IIb) in the heterodimers was human, rat, or a rat-human chimera. These results indicate that the sequences determining the sensitivity of alpha(IIb)beta(3) to RGD-containing peptides are located in the third and fourth amino-terminal repeats of alpha(IIb). Because RGDS binds to both human and rat alpha(IIb)beta(3), the results suggest that differences in RGDS sensitivity result from differences in the allosteric changes induced in these repeats following RGDS binding.


Asunto(s)
Plaquetas/metabolismo , Fibrinógeno/química , Fibrinógeno/metabolismo , Oligopéptidos/química , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/química , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Adenosina Difosfato/metabolismo , Sitio Alostérico , Animales , Linfocitos B/metabolismo , Células CHO , Carcinógenos , Adhesión Celular , Cricetinae , ADN Complementario/metabolismo , Ditiotreitol/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Ligandos , Agregación Plaquetaria , Unión Proteica , Estructura Terciaria de Proteína , Ratas , Proteínas Recombinantes de Fusión/química , Acetato de Tetradecanoilforbol , Transfección
19.
Blood ; 96(13): 4142-51, 2000 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-11110685

RESUMEN

The role of the chemokine binding stromal-derived factor 1 (SDF-1) in normal human megakaryopoiesis at the cellular and molecular levels and its comparison with that of thrombopoietin (TPO) have not been determined. In this study it was found that SDF-1, unlike TPO, does not stimulate alpha(IIb)beta(3)(+) cell proliferation or differentiation or have an antiapoptotic effect. However, it does induce chemotaxis, trans-Matrigel migration, and secretion of matrix metalloproteinase 9 (MMP-9) and vascular endothelial growth factor (VEGF) by these cells, and both SDF-1 and TPO increase the adhesion of alpha(IIb)beta(3)(+) cells to fibrinogen and vitronectin. Investigating the intracellular signaling pathways induced by SDF-1 and TPO revealed some overlapping patterns of protein phosphorylation/activation (mitogen-activated protein kinase [MAPK] p42/44, MAPK p38, and AKT [protein kinase B]) and some that were distinct for TPO (eg, JAK-STAT) and for SDF-1 (eg, NF-kappa B). It was also found that though inhibition of phosphatidyl-inositol 3-kinase (PI-3K) by LY294002 in alpha(IIb)beta(3)(+) cells induced apoptosis and inhibited chemotaxis adhesion and the secretion of MMP-9 and VEGF, the inhibition of MAPK p42/44 (by the MEK inhibitor U0126) had no effect on the survival, proliferation, and migration of these cells. Hence, it is suggested that the proliferative effect of TPO is more related to activation of the JAK-STAT pathway (unique to TPO), and the PI-3K-AKT axis is differentially involved in TPO- and SDF-1-dependent signaling. Accordingly, PI-3K is involved in TPO-mediated inhibition of apoptosis, TPO- and SDF-1-regulated adhesion to fibrinogen and vitronectin, and SDF-1-mediated migration. This study expands the understanding of the role of SDF-1 and TPO in normal human megakaryopoiesis and indicates the molecular basis of the observed differences in cellular responses. (Blood. 2000;96:4142-4151)


Asunto(s)
Quimiocinas CXC/fisiología , Megacariocitos/citología , Trombopoyetina/fisiología , Adulto , Apoptosis/efectos de los fármacos , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Caspasa 3 , Caspasas/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Quimiocina CXCL12 , Quimiocinas CXC/farmacología , Quimiotaxis/efectos de los fármacos , Cromonas/farmacología , Colágeno , Combinación de Medicamentos , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Fibrinógeno/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Transporte Iónico/efectos de los fármacos , Laminina , Linfocinas/biosíntesis , Linfocinas/genética , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Megacariocitos/efectos de los fármacos , Metaloendopeptidasas/biosíntesis , Metaloendopeptidasas/genética , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/fisiología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteoglicanos , Proteínas Recombinantes/farmacología , Trombopoyetina/farmacología , Factores de Transcripción/metabolismo , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular , Vitronectina/metabolismo
20.
Proc Natl Acad Sci U S A ; 97(18): 9984-9, 2000 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-10954748

RESUMEN

Heterotrimeric G proteins mediate the earliest step in cell responses to external events by linking cell surface receptors to intracellular signaling pathways. G(z) is a member of the G(i) family of G proteins that is prominently expressed in platelets and brain. Here, we show that deletion of the alpha subunit of G(z) in mice: (i) impairs platelet aggregation by preventing the inhibition of cAMP formation normally seen at physiologic concentrations of epinephrine, and (ii) causes the mice to be more resistant to fatal thromboembolism. Loss of G(zalpha) also results in greatly exaggerated responses to cocaine, reduces the analgesic effects of morphine, and abolishes the effects of widely used antidepressant drugs that act as catecholamine reuptake inhibitors. These changes occur despite the presence of other G(ialpha) family members in the same cells and are not accompanied by detectable compensatory changes in the level of expression of other G protein subunits. Therefore, these results provide insights into receptor selectivity among G proteins and a model for understanding platelet function and the effects of psychoactive drugs.


Asunto(s)
Plaquetas/fisiología , Encéfalo/fisiología , Subunidades alfa de la Proteína de Unión al GTP , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Activación Plaquetaria/fisiología , Psicotrópicos/farmacología , Animales , Plaquetas/efectos de los fármacos , Encéfalo/efectos de los fármacos , Cocaína/farmacología , Colágeno/farmacología , AMP Cíclico/sangre , Epinefrina/farmacología , Proteínas de Unión al GTP Heterotriméricas/deficiencia , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Masculino , Ratones , Ratones Noqueados , Morfina/farmacología , Actividad Motora , Agregación Plaquetaria , Transducción de Señal , Tromboembolia/genética , Tromboembolia/fisiopatología , Tromboembolia/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...