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 Adv ; 6(17): 5100-5112, 2022 09 13.
Artigo em Inglês | MEDLINE | ID: mdl-35839075

RESUMO

Transcription factor RUNX1 is a master regulator of hematopoiesis and megakaryopoiesis. RUNX1 haplodeficiency (RHD) is associated with thrombocytopenia and platelet granule deficiencies and dysfunction. Platelet profiling of our study patient with RHD showed decreased expression of RAB31, a small GTPase whose cell biology in megakaryocytes (MKs)/platelets is unknown. Platelet RAB31 messenger RNA was decreased in the index patient and in 2 additional patients with RHD. Promoter-reporter studies using phorbol 12-myristate 13-acetate-treated megakaryocytic human erythroleukemia cells revealed that RUNX1 regulates RAB31 via binding to its promoter. We investigated RUNX1 and RAB31 roles in endosomal dynamics using immunofluorescence staining for markers of early endosomes (EEs; early endosomal autoantigen 1) and late endosomes (CD63)/multivesicular bodies. Downregulation of RUNX1 or RAB31 (by small interfering RNA or CRISPR/Cas9) showed a striking enlargement of EEs, partially reversed by RAB31 reconstitution. This EE defect was observed in MKs differentiated from a patient-derived induced pluripotent stem cell line (RHD-iMKs). Studies using immunofluorescence staining showed that trafficking of 3 proteins with distinct roles (von Willebrand factor [VWF], a protein trafficked to α-granules; epidermal growth factor receptor; and mannose-6-phosphate) was impaired at the level of EE on downregulation of RAB31 or RUNX1. There was loss of plasma membrane VWF in RUNX1- and RAB31-deficient megakaryocytic human erythroleukemia cells and RHD-iMKs. These studies provide evidence that RAB31 is downregulated in RHD and regulates megakaryocytic vesicle trafficking of 3 major proteins with diverse biological roles. EE defect and impaired vesicle trafficking is a potential mechanism for the α-granule defects observed in RUNX1 deficiency.


Assuntos
Leucemia Eritroblástica Aguda , Megacariócitos , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Receptores ErbB/metabolismo , Humanos , Megacariócitos/metabolismo , Proteínas rab de Ligação ao GTP/genética , Proteínas rab de Ligação ao GTP/metabolismo , Fator de von Willebrand/metabolismo
2.
J Neuroimmune Pharmacol ; 14(4): 578-594, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31512166

RESUMO

Virus-induced diseases or neurological complications are huge socio-economic burden to human health globally. The complexity of viral-mediated CNS pathology is exacerbated by reemergence of new pathogenic neurotropic viruses of high public relevance. Although the central nervous system is considered as an immune privileged organ and is mainly protected by barrier system, there are a vast majority of neurotropic viruses capable of gaining access and cause diseases. Despite continued growth of the patient population and a number of treatment strategies, there is no successful viral specific therapy available for viral induced CNS diseases. Therefore, there is an urgent need for a clear alternative treatment strategy that can effectively target neurotropic viruses of DNA or RNA genome. To address this need, rapidly growing gene editing technology based on CRISPR/Cas9, provides unprecedented control over viral genome editing and will be an effective, highly specific and versatile tool for targeting CNS viral infection. In this review, we discuss the application of this system to control CNS viral infection and associated neurological disorders and future prospects. Graphical Abstract CRISPR/Cas9 technology as agent control over CNS viral infection.


Assuntos
Proteína 9 Associada à CRISPR/genética , Sistemas CRISPR-Cas/genética , Doenças do Sistema Nervoso Central/genética , Doenças do Sistema Nervoso Central/terapia , Edição de Genes/tendências , Terapia Genética/tendências , Animais , Proteína 9 Associada à CRISPR/metabolismo , Doenças do Sistema Nervoso Central/metabolismo , Edição de Genes/métodos , Terapia Genética/métodos , Humanos
3.
Blood Adv ; 2(7): 797-806, 2018 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-29632235

RESUMO

Patients with RUNX1 haplodeficiency have thrombocytopenia, platelet dysfunction, and deficiencies of α-granules and dense granules. Platelet expression profiling of a patient with a heterozygous RUNX1 mutation (c.969-323G>T) revealed decreased RAB1B, which encodes a small G protein. RAB GTPases regulate vesicle trafficking, and RAB1B is implicated in endoplasmic reticulum (ER)-to-Golgi transport in nonhematopoietic cells, but its role in megakaryocytes (MK) is unknown. We addressed the hypothesis that RAB1B is a transcriptional target of RUNX1 and that RAB1B regulates ER-to-Golgi transport in MK cells. Chromatin immunoprecipitation studies and electrophoretic mobility shift assay using phorbol 12-myristate 13-acetate (PMA)-treated human erythroleukemia cells revealed RUNX1 binding to RAB1B promoter region RUNX1 consensus sites, and their mutation reduced the promoter activity. RAB1B promoter activity and protein expression were inhibited by RUNX1 siRNA and enhanced by RUNX1 overexpression. These indicate that RAB1B is a direct RUNX1 target, providing a mechanism for decreased RAB1B in patient platelets. Vesicle trafficking from ER to Golgi in PMA-treated human erythroleukemia cells was impaired along with Golgi disruption on siRNA downregulation of RUNX1 or RAB1B. The effects of RUNX1 knockdown were reversed by RAB1B reconstitution. Trafficking of von Willebrand factor (vWF), an α-granule MK synthesized protein, was impaired with RUNX1 or RAB1B downregulation and reconstituted by ectopic RAB1B expression. Platelet vWF was decreased in patients with RUNX1 mutations. Thus, ER-to-Golgi transport, an early critical step in protein trafficking to granules, is impaired in megakaryocytic cells on RUNX1 downregulation, secondary to decreased RAB1B expression. Impaired RAB1B mediated ER-to-Golgi transport contributes to platelet α-granule defects in RUNX1 haplodeficiency.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Megacariócitos/química , Proteínas rab1 de Ligação ao GTP/metabolismo , Plaquetas/química , Estudos de Casos e Controles , Células Cultivadas , Criança , Pré-Escolar , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Retículo Endoplasmático/metabolismo , Feminino , Complexo de Golgi/metabolismo , Humanos , Leucemia Eritroblástica Aguda/patologia , Masculino , Regiões Promotoras Genéticas , Ligação Proteica , Transporte Proteico , Proteínas rab1 de Ligação ao GTP/genética , Fator de von Willebrand/metabolismo
4.
EBioMedicine ; 11: 157-164, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27566955

RESUMO

Aspirin prevents cardiovascular disease and colon cancer; however aspirin's inhibition of platelet COX-1 only partially explains its diverse effects. We previously identified an aspirin response signature (ARS) in blood consisting of 62 co-expressed transcripts that correlated with aspirin's effects on platelets and myocardial infarction (MI). Here we report that 60% of ARS transcripts are regulated by RUNX1 - a hematopoietic transcription factor - and 48% of ARS gene promoters contain a RUNX1 binding site. Megakaryocytic cells exposed to aspirin and its metabolite (salicylic acid, a weak COX-1 inhibitor) showed up regulation in the RUNX1 P1 isoform and MYL9, which is transcriptionally regulated by RUNX1. In human subjects, RUNX1 P1 expression in blood and RUNX1-regulated platelet proteins, including MYL9, were aspirin-responsive and associated with platelet function. In cardiovascular disease patients RUNX1 P1 expression was associated with death or MI. RUNX1 acts as a tumor suppressor gene in gastrointestinal malignancies. We show that RUNX1 P1 expression is associated with colon cancer free survival suggesting a role for RUNX1 in aspirin's protective effect in colon cancer. Our studies reveal an effect of aspirin on RUNX1 and gene expression that may additionally explain aspirin's effects in cardiovascular disease and cancer.


Assuntos
Aspirina/farmacologia , Doenças Cardiovasculares/genética , Neoplasias do Colo/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Predisposição Genética para Doença , Aspirina/uso terapêutico , Plaquetas/metabolismo , Doenças Cardiovasculares/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/mortalidade , Neoplasias do Colo/patologia , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Perfilação da Expressão Gênica , Humanos , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Estadiamento de Neoplasias , Farmacogenética , Prognóstico
5.
Thromb Res ; 134(3): 704-10, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25042561

RESUMO

INTRODUCTION: Diabetes mellitus (DM) is a prothrombotic and proinflammatory state. Hyperglycemia (HG) is encountered even in patients without DM. We have shown that combined HG and hyperinsulinemia (HI) in healthy non-diabetic subjects increased circulating tissue factor (TF) and thrombin generation. To understand the changes in platelet and monocyte pathways induced by combined HG and HI in healthy non-diabetic state, we performed whole genome expression profiling of leukocyte-depleted platelets and monocytes before and after 24 hours of combined HG (glucose ~200mg/dL) and HI by glucose infusion clamp in a healthy non-diabetic subject. RESULTS: We defined time-dependent differential mRNA expression (24 versus 0 hour fold change (FC) ≥ 2) common to platelets and monocytes. Ingenuity Pathways Analysis revealed alterations in canonical insulin receptor signaling and coagulation pathways. A preliminary group of 9 differentially expressed genes was selected for qRT-PCR confirmation. Platelet 24 hour sample was compared to the 0 hour sample plus 4 controls. Five transcripts in platelets and 6 in monocytes were confirmed. Platelet GSK3B and PTPN1 were upregulated, and STXBP4 was downregulated in insulin signaling, and F3 and TFPI were upregulated in coagulation pathways. Monocyte, PIK3C3, PTPN11 and TFPI were downregulated. Platelet GSKß3 and PTPN11 protein and TF antigen in platelets and monocytes was increased. CONCLUSIONS: Even in non-diabetic state, HG+HI for 24 hours induces changes in platelets and monocytes. They suggest downregulation of insulin signaling and upregulation of TF. Further studies are needed to elucidate cellular alterations leading to the prothrombotic and proinflammatory state in DM.


Assuntos
Coagulação Sanguínea , Plaquetas/metabolismo , Hiperglicemia/sangue , Hiperinsulinismo/sangue , Insulina/sangue , Transdução de Sinais , Adulto , Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/genética , Fatores de Coagulação Sanguínea/metabolismo , Análise por Conglomerados , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Marcadores Genéticos , Voluntários Saudáveis , Humanos , Hiperglicemia/diagnóstico , Hiperglicemia/genética , Hiperinsulinismo/diagnóstico , Hiperinsulinismo/genética , Masculino , Monócitos/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/sangue , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/genética , Fatores de Tempo
6.
Arterioscler Thromb Vasc Biol ; 31(4): 921-7, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21252065

RESUMO

OBJECTIVE: Mutations in the hematopoietic transcription factor RUNX1 cause thrombocytopenia and impaired platelet function. In a patient with a heterozygous mutation in RUNX1, we have described decreased platelet pleckstrin phosphorylation and protein kinase C- (PKC-, gene PRKCQ) associated with thrombocytopenia, impaired platelet aggregation, and dense granule secretion. Little is known regarding regulation of PKC- in megakaryocytes and platelets. We have addressed the hypothesis that PRKCQ is a direct transcriptional target of RUNX1. METHODS AND RESULTS: In a chromatin immunoprecipitation assay using megakaryocytic cells, there was RUNX1 binding in vivo to PRKCQ promoter region -1225 to -1056 bp containing a RUNX1 consensus site ACCGCA at -1088 to -1069 bp; an electrophoretic mobility shift assay showed RUNX1 binding to the specific site. In RUNX1 overexpression studies, PKC- protein expression and promoter activity were enhanced; mutation of RUNX1 site showed decreased activity even with RUNX1 overexpression. Lastly, PRKCQ promoter activity and PKC- protein were decreased by short interfering RNA knockdown of RUNX1. CONCLUSIONS: Our results provide the first evidence that PRKCQ is regulated at the transcriptional level by RUNX1 in megakaryocytic cells and a mechanism for PKC- deficiency associated with RUNX1 haplodeficiency.


Assuntos
Plaquetas/enzimologia , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Regulação Enzimológica da Expressão Gênica , Isoenzimas/genética , Megacariócitos/enzimologia , Mutação , Proteína Quinase C/genética , Trombocitopenia/genética , Transcrição Gênica , Sítios de Ligação , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Sequência Consenso , Subunidade alfa 2 de Fator de Ligação ao Core/sangue , Ensaio de Desvio de Mobilidade Eletroforética , Genes Reporter , Humanos , Isoenzimas/sangue , Isoenzimas/deficiência , Masculino , Regiões Promotoras Genéticas , Proteína Quinase C/sangue , Proteína Quinase C/deficiência , Proteína Quinase C-theta , Interferência de RNA , Proteínas Recombinantes/metabolismo , Trombocitopenia/sangue , Trombocitopenia/enzimologia , Transfecção , Adulto Jovem
7.
Blood ; 116(26): 6037-45, 2010 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-20876458

RESUMO

Mutations in transcription factor RUNX1 are associated with familial platelet disorder, thrombocytopenia, and predisposition to leukemia. We have described a patient with thrombocytopenia and impaired agonist-induced platelet aggregation, secretion, and glycoprotein (GP) IIb-IIIa activation, associated with a RUNX1 mutation. Platelet myosin light chain (MLC) phosphorylation and transcript levels of its gene MYL9 were decreased. Myosin IIA and MLC phosphorylation are important in platelet responses to activation and regulate thrombopoiesis by a negative regulatory effect on premature proplatelet formation. We addressed the hypothesis that MYL9 is a transcriptional target of RUNX1. Chromatin immunoprecipitation (ChIP) using megakaryocytic cells revealed RUNX1 binding to MYL9 promoter region -729/-542 basepairs (bp), which contains 4 RUNX1 sites. Electrophoretic mobility shift assay showed RUNX1 binding to each site. In transient ChIP assay, mutation of these sites abolished binding of RUNX1 to MYL9 promoter construct. In reporter gene assays, deletion of each RUNX1 site reduced activity. MYL9 expression was inhibited by RUNX1 short interfering RNA (siRNA) and enhanced by RUNX1 overexpression. RUNX1 siRNA decreased cell spreading on collagen and fibrinogen. Our results constitute the first evidence that the MYL9 gene is a direct target of RUNX1 and provide a mechanism for decreased platelet MYL9 expression, MLC phosphorylation, thrombocytopenia, and platelet dysfunction associated with RUNX1 mutations.


Assuntos
Transtornos Plaquetários/genética , Plaquetas/metabolismo , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica , Cadeias Leves de Miosina/genética , Regiões Promotoras Genéticas/genética , Trombocitopenia/genética , Sequência de Bases , Transtornos Plaquetários/metabolismo , Transtornos Plaquetários/patologia , Células Cultivadas , Subunidade alfa 2 de Fator de Ligação ao Core/antagonistas & inibidores , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Humanos , Dados de Sequência Molecular , Cadeias Leves de Miosina/metabolismo , Agregação Plaquetária , RNA Interferente Pequeno/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia
8.
Blood ; 115(15): 3128-35, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20181616

RESUMO

Haploinsufficiency of RUNX1 (also known as CBFA2/AML1) is associated with familial thrombocytopenia, platelet dysfunction, and predisposition to acute leukemia. We have reported on a patient with thrombocytopenia and impaired agonist-induced aggregation, secretion, and protein phosphorylation associated with a RUNX1 mutation. Expression profiling of platelets revealed approximately 5-fold decreased expression of 12-lipoxygenase (12-LO, gene ALOX12), which catalyzes 12-hydroxyeicosatetraenoic acid production from arachidonic acid. We hypothesized that ALOX12 is a direct transcriptional target gene of RUNX1. In present studies, agonist-induced platelet 12-HETE production was decreased in the patient. Four RUNX1 consensus sites were identified in the 2-kb promoter region of ALOX12 (at -1498, -1491, -708, -526 from ATG). In luciferase reporter studies in human erythroleukemia cells, mutation of each site decreased activity; overexpression of RUNX1 up-regulated promoter activity, which was abolished by mutation of RUNX1 sites. Gel shift studies, including with recombinant protein, revealed RUNX1 binding to each site. Chromatin immunoprecipitation revealed in vivo RUNX1 binding in the region of interest. siRNA knockdown of RUNX1 decreased RUNX1 and 12-LO proteins. ALOX12 is a direct transcriptional target of RUNX1. Our studies provide further proof of principle that platelet expression profiling can elucidate novel alterations in platelets with inherited dysfunction.


Assuntos
Araquidonato 12-Lipoxigenase/genética , Plaquetas/enzimologia , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Haploidia , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico/biossíntese , Araquidonato 12-Lipoxigenase/metabolismo , Ácido Araquidônico/farmacologia , Sequência de Bases , Sítios de Ligação , Plaquetas/efeitos dos fármacos , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Sequência Consenso , Ensaio de Desvio de Mobilidade Eletroforética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Genes Reporter , Humanos , Leucemia Eritroblástica Aguda/enzimologia , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/patologia , Luciferases/metabolismo , Dados de Sequência Molecular , Ativação Plaquetária/efeitos dos fármacos , Regiões Promotoras Genéticas/genética , Ligação Proteica/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Trombina/farmacologia
9.
Thromb Haemost ; 100(5): 821-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18989526

RESUMO

Galphaq plays a major role in platelet signal transduction, but little is known regarding its transcriptional regulation. We have reported that Galphaq is upregulated during phorbol 12-myristate 13-acetate (PMA)-induced megakaryocytic transformation of human erythroleukemia (HEL) cells and regulated by EGR-1, an early growth transcription factor. These studies focused on the initial 238 bp of the 5' upstream region of the Galphaq gene. In the present studies we characterize a minimal region -1042/-1037 bp from ATG in the 5' upstream of the Galphaq promoter that is associated with PMA responsiveness. In luciferase reporter gene studies in HEL cells, Galphaq 5' upstream promoter sequence -1042/-1 showed an about four-fold increased activity in PMA-treated compared to untreated cells. Deletion of 6-nt -1042/-1037 eliminated the difference. Gel-shift studies on Galphaq probe (-1042/-1012 bp) revealed binding of EGR-1 with PMA-treated but not untreated nuclear extracts, and this was dependent on the sequence -1042/-1037. Silencing of endogenous EGR-1 inhibited Galphaq induction by PMA. MEK/ERK inhibitor U0126 blocked PMA effect on promoter activity of the -1042/-1 construct. In conclusion, EGR-1 binding to sequence -1042/-1037 bp in Galphaq promoter mediates the induction of Galphaq gene by PMA via the MEK/ERK signaling pathway. These studies provide the first evidence of a PMA-responsive element in Galphaq promoter, and new insights into regulation of Galphaq gene by EGR-1.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Megacariócitos/efeitos dos fármacos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Elementos de Resposta/efeitos dos fármacos , Acetato de Tetradecanoilforbol/farmacologia , Região 5'-Flanqueadora/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proteína 1 de Resposta de Crescimento Precoce/genética , Ensaio de Desvio de Mobilidade Eletroforética , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/genética , Genes Reporter , Humanos , Megacariócitos/enzimologia , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Oligonucleotídeos Antissenso/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima
10.
Thromb Haemost ; 96(6): 738-43, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17139367

RESUMO

Tissue factor (TF) is the physiological initiating mechanism for blood coagulation. Platelets play an important role in monocyte TF expression, thrombosis and inflammation. Aspirin, clopidogrel and cilostazol, which inhibit platelet responses by different mechanisms, are widely used in patients with arterial diseases. We tested the hypothesis that platelet-inhibiting agents inhibit the levels of circulating TF procoagulant activity (TF-PCA) in patients with peripheral arterial disease (PAD). Twenty-six patients with lower extremity PAD, average age 65.9 +/- 8.4 years (mean +/- SEM), were studied at baseline and following sequential two-week treatment regimens with aspirin (325 mg daily), clopidogrel (75 mg daily) or a phosphodiesterase inhibitor cilostazol (100 mg twice daily) singly, and with each possible combination of these agents. Circulating TF-PCA in whole blood, and plasma factor VIIa, prothrombin fragment F1.2, thrombin-antithrombin complexes (TAT), and P-selectin were measured. Baseline TF-PCA levels in the patients were elevated (131 +/- 19 U/ml) compared to control subjects (23 +/- 2, p < 0.0001). TF-PCA levels declined following treatment with clopidogrel alone, and with combinations of clopidogrel with aspirin or cilostazol, with the lowest levels being with the triple-drug combination. Plasma P-selectin declined in all treatment groups. No changes were noted in plasma factor VIIa, F1.2 or TAT. In conclusion, treatment of PAD patients with antiplatelet agents decreases circulating TF, a molecule with prothrombotic and proinflammatory effects. These findings suggest an unrecognized mechanism, beyond inhibiting aggregation responses, for the efficacy of antiplatelet drugs in patients with arterial diseases.


Assuntos
Aspirina/uso terapêutico , Doenças Vasculares Periféricas/tratamento farmacológico , Inibidores da Agregação Plaquetária/uso terapêutico , Tetrazóis/uso terapêutico , Tromboplastina/metabolismo , Ticlopidina/análogos & derivados , Idoso , Idoso de 80 Anos ou mais , Antitrombina III , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Proteína C-Reativa/metabolismo , Ligante de CD40/sangue , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cilostazol , Clopidogrel , Regulação para Baixo/efeitos dos fármacos , Quimioterapia Combinada , Fator VIIa/metabolismo , Humanos , Pessoa de Meia-Idade , Selectina-P/sangue , Fragmentos de Peptídeos/sangue , Peptídeo Hidrolases/sangue , Doenças Vasculares Periféricas/sangue , Projetos Piloto , Inibidores da Agregação Plaquetária/farmacologia , Estudos Prospectivos , Protrombina , Ticlopidina/uso terapêutico
11.
Semin Thromb Hemost ; 30(5): 525-35, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15497095

RESUMO

In the majority of patients with an inherited abnormality in platelet function and a bleeding diathesis, the underlying platelet molecular mechanisms are unknown. The usually considered entities, such as thrombasthenia, the Bernard-Soulier syndrome, and storage pool deficiency, occur in a small proportion of patients. A substantial number of patients present with decreased aggregation and secretion of dense granule contents upon activation, and are lumped in the category of primary secretion defects or platelet activation defects. Evidence is now available that defects in platelet signaling mechanisms may be the basis for the platelet dysfunction in some of these patients. This evidence is presented here. If the key components in signal transduction are the surface receptors, the G-proteins, and the effectors, evidence now exists for specific human platelet abnormalities at each of these levels. There is a pressing need for a concerted effort to delineate the molecular mechanisms in the large group of patients with impaired platelet function who represent an untapped reservoir of new information into normal platelet function.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Ácido Araquidônico/metabolismo , Síndrome de Bernard-Soulier/sangue , Citoesqueleto/metabolismo , Ativação Enzimática , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Humanos , Isoenzimas/deficiência , Modelos Biológicos , Fosfolipase C beta , Deficiência do Pool Plaquetário/sangue , Proteína Quinase C/metabolismo , Transdução de Sinais , Trombastenia/sangue , Tromboxano A2/metabolismo , Tromboxano-A Sintase/metabolismo , Fosfolipases Tipo C/deficiência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...