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.
Arterioscler Thromb Vasc Biol ; 43(10): 2042-2057, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37589138

RESUMO

BACKGROUND: ADP-induced platelet activation leads to cell surface expression of several proteins, including TF (tissue factor). The role of ADP receptors in platelet TF modulation is still unknown. We aimed to assess the (1) involvement of P2Y1 and P2Y12 receptors in ADP-induced TF exposure; (2) modulation of TFpos-platelets in anti-P2Y12-treated patients with coronary artery disease. Based on the obtained results, we revisited the intracellular localization of TF in platelets. METHODS: The effects of P2Y1 or P2Y12 antagonists on ADP-induced TF expression and activity were analyzed in vitro by flow cytometry and thrombin generation assay in blood from healthy subjects, P2Y12-/-, and patients with gray platelet syndrome. Ex vivo, P2Y12 inhibition of TF expression by clopidogrel/prasugrel/ticagrelor, assessed by VASP (vasodilator-stimulated phosphoprotein) platelet reactivity index, was investigated in coronary artery disease (n=238). Inhibition of open canalicular system externalization and electron microscopy (TEM) were used for TF localization. RESULTS: In blood from healthy subjects, stimulated in vitro by ADP, the percentage of TFpos-platelets (17.3±5.5%) was significantly reduced in a concentration-dependent manner by P2Y12 inhibition only (-81.7±9.5% with 100 nM AR-C69931MX). In coronary artery disease, inhibition of P2Y12 is paralleled by reduction of ADP-induced platelet TF expression (VASP platelet reactivity index: 17.9±11%, 20.9±11.3%, 40.3±13%; TFpos-platelets: 10.5±4.8%, 9.8±5.9%, 13.6±6.3%, in prasugrel/ticagrelor/clopidogrel-treated patients, respectively). Despite this, 15% of clopidogrel good responders had a level of TFpos-platelets similar to the poor-responder group. Indeed, a stronger P2Y12 inhibition (130-fold) is required to inhibit TF than VASP. Thus, a VASP platelet reactivity index <20% (as in prasugrel/ticagrelor-treated patients) identifies patients with TFpos-platelets <20% (92% sensitivity). Finally, colchicine impaired in vitro ADP-induced TF expression but not α-granule release, suggesting that TF is open canalicular system stored as confirmed by TEM and platelet analysis of patients with gray platelet syndrome. CONCLUSIONS: Data show that TF expression is regulated by P2Y12 and not P2Y1; P2Y12 antagonists downregulate the percentage of TFpos-platelets. In clopidogrel good-responder patients, assessment of TFpos-platelets highlights those with residual platelet reactivity. TF is stored in open canalicular system, and its membrane exposure upon activation is prevented by colchicine.


Assuntos
Doença da Artéria Coronariana , Síndrome da Plaqueta Cinza , Humanos , Plaquetas/metabolismo , Clopidogrel/farmacologia , Doença da Artéria Coronariana/metabolismo , Síndrome da Plaqueta Cinza/metabolismo , Agregação Plaquetária , Inibidores da Agregação Plaquetária/farmacologia , Inibidores da Agregação Plaquetária/uso terapêutico , Inibidores da Agregação Plaquetária/metabolismo , Testes de Função Plaquetária/métodos , Cloridrato de Prasugrel/metabolismo , Cloridrato de Prasugrel/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y12 , Tromboplastina/metabolismo , Ticagrelor
2.
Transfusion ; 59(1): 32-38, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30394544

RESUMO

BACKGROUND: Beyond their role in hemostasis and thrombosis, platelets are also important mediators of inflammation by the release of hundreds of factors stored in their α-granules. Mutations in Nbeal2 cause gray platelet syndrome (GPS), characterized by the lack of platelet α-granules. This study aims to evaluate the immunological (proinflammatory) effects of platelet α-granules. STUDY DESIGN AND METHODS: We performed an experiment using Nbeal2-/- mice, the mouse model of GPS. Systemic inflammation was induced by intravenous injection of lipopolysaccharide (LPS). Inflammatory response was assessed by quantification of inflammatory soluble factors and platelet biological response modifiers. RESULTS: The lack of Nbeal2 (in Nbeal2 -/- mice, compared with controls) significantly reduced the recruitment of circulating neutrophils and monocytes. Moreover, after LPS injection, there was a significant increase in neutrophil and monocyte counts in control animals, compared with Nbeal2 -/- mice. The control of inflammation, evaluated by the production of anti-inflammatory cytokines, appeared to be greater in Nbeal2-/- mice compared with controls. Conversely, the production of certain inflammatory-soluble mediators known to characterize normal platelet secretion, such as soluble CD40 ligand (sCD40L), was decreased under experimental inflammation in Nbeal2 -/- mice. CONCLUSIONS: These results show that α-granules play a direct role in platelet-mediated inflammation balance, confirming the need to further investigate platelet-associated inflammatory pathophysiology and inflammatory adverse events related to blood transfusion.


Assuntos
Proteínas Sanguíneas/metabolismo , Síndrome da Plaqueta Cinza/imunologia , Lipopolissacarídeos/toxicidade , Animais , Proteínas Sanguíneas/genética , Ligante de CD40/genética , Ligante de CD40/metabolismo , Modelos Animais de Doenças , Síndrome da Plaqueta Cinza/genética , Síndrome da Plaqueta Cinza/metabolismo , Imunoensaio , Inflamação/induzido quimicamente , Inflamação/genética , Inflamação/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação/genética
3.
Biosci Rep ; 38(5)2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30104399

RESUMO

Platelets respond to vascular injury via surface receptor stimulation and signaling events to trigger aggregation, procoagulant activation, and granule secretion during hemostasis, thrombosis, and vascular remodeling. Platelets contain three major types of secretory granules including dense granules (or δ-granules, DGs), α-granules (AGs), and lysosomes. The contents of platelet granules are specific. Platelet DGs store polyphosphate and small molecules such as ADP, ATP, Ca2+, and serotonin, while AGs package most of the proteins that platelets release. The platelet DGs and AGs are regarded as being budded from the endosomes and the trans-Golgi network (TGN), respectively, and then matured from multivesicular bodies (MVBs). However, the sorting machineries between DGs and AGs are different. Inherited platelet disorders are associated with deficiency of DGs and AGs, leading to bleeding diathesis in patients with Hermansky-Pudlak syndrome (HPS), gray platelet syndrome (GPS), and arthrogryposis, renal dysfunction, and cholestasis syndrome (ARC). Here, we reviewed the current understanding about how DGs differ from AGs in structure, biogenesis, and function. In particular, we focus on the sorting machineries that are involved in the formation of these two types of granules to provide insights into their diverse biological functions.


Assuntos
Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Corpos Multivesiculares/metabolismo , Vesículas Secretórias/metabolismo , Artrogripose/metabolismo , Artrogripose/patologia , Plaquetas/patologia , Colestase/metabolismo , Colestase/patologia , Grânulos Citoplasmáticos/genética , Endossomos/metabolismo , Síndrome da Plaqueta Cinza/metabolismo , Síndrome da Plaqueta Cinza/patologia , Síndrome de Hermanski-Pudlak/metabolismo , Síndrome de Hermanski-Pudlak/patologia , Humanos , Lisossomos/metabolismo , Insuficiência Renal/metabolismo , Insuficiência Renal/patologia , Vesículas Secretórias/genética , Rede trans-Golgi/genética
4.
Arterioscler Thromb Vasc Biol ; 38(8): 1772-1784, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29930006

RESUMO

Objective- Nbeal2-/- mice, a model of human gray platelet syndrome, have reduced neutrophil granularity and impaired host defense against systemic Staphylococcus aureus infection. We here aimed to study the role of Nbeal2 deficiency in both leukocytes and platelets during gram-negative pneumonia and sepsis. Approach and Results- We studied the role of Nbeal2 in platelets and leukocytes during murine pneumonia and sepsis by Klebsiella pneumoniae. Apart from platelet α-granule deficiency and reduced neutrophil granularity, also monocyte granularity was reduced in Nbeal2-/- mice, whereas plasma levels of MPO (myeloperoxidase), elastase, NGAL (neutrophil gelatinase-associated lipocalin), and MMP-9 (matrix metalloproteinase 9), and leukocyte CD11b expression were increased. Nbeal2-/- leukocytes showed unaltered in vitro antibacterial response and phagocytosis capacity against Klebsiella, and unchanged reactive nitrogen species and cytokine production. Also during Klebsiella pneumonia and sepsis, Nbeal2-/- mice had similar bacterial growth in lung and distant body sites, with enhanced leukocyte migration to the bronchoalveolar space. Despite similar infection-induced inflammation, organ damage was increased in Nbeal2-/- mice, which was also seen during endotoxemia. Platelet-specific Nbeal2 deficiency did not influence leukocyte functions, indicating that Nbeal2 directly modifies leukocytes. Transfusion of Nbeal2-/- but not of Nbeal2+/+ platelets into thrombocytopenic mice was associated with bleeding in the lung but similar host defense, pointing at a role for platelet α-granules in maintaining vascular integrity but not host defense during Klebsiella pneumosepsis. Conclusions- These data show that Nbeal2 deficiency-resulting in gray platelet syndrome-affects platelets, neutrophils, and monocytes, with intact host defense but increased organ damage during gram-negative pneumosepsis.


Assuntos
Plaquetas/metabolismo , Proteínas Sanguíneas/deficiência , Síndrome da Plaqueta Cinza/metabolismo , Infecções por Klebsiella/metabolismo , Klebsiella pneumoniae/patogenicidade , Insuficiência de Múltiplos Órgãos/metabolismo , Pneumonia Bacteriana/metabolismo , Sepse/metabolismo , Animais , Plaquetas/microbiologia , Proteínas Sanguíneas/genética , Antígeno CD11b/sangue , Modelos Animais de Doenças , Feminino , Síndrome da Plaqueta Cinza/sangue , Síndrome da Plaqueta Cinza/genética , Interações Hospedeiro-Patógeno , Infecções por Klebsiella/sangue , Infecções por Klebsiella/genética , Infecções por Klebsiella/microbiologia , Klebsiella pneumoniae/crescimento & desenvolvimento , Lipocalina-2/sangue , Masculino , Metaloproteinase 9 da Matriz/sangue , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Monócitos/microbiologia , Insuficiência de Múltiplos Órgãos/sangue , Insuficiência de Múltiplos Órgãos/genética , Insuficiência de Múltiplos Órgãos/microbiologia , Neutrófilos/metabolismo , Neutrófilos/microbiologia , Elastase Pancreática/sangue , Peroxidase/sangue , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Transfusão de Plaquetas , Pneumonia Bacteriana/sangue , Pneumonia Bacteriana/genética , Pneumonia Bacteriana/microbiologia , Sepse/sangue , Sepse/genética , Sepse/microbiologia
5.
Platelets ; 29(6): 632-635, 2018 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-29869935

RESUMO

Homozygosity/compound heterozygosity for loss of function mutations in neurobeachin-like 2 (NBEAL2) is causative for Gray platelet syndrome (GPS; MIM #139090), characterized by thrombocytopenia and large platelets lacking α-granules and cargo. Most GPS-associated NBEAL2 mutations generate nonsense codons; frameshifts causing premature translation termination and/or changes in mRNA splicing have also been observed. Data regarding NBEAL2 protein expression in GPS patients is limited. We observed absence of NBEAL2 in platelets from GPS patients with 3 different genotypes, and reduced/truncated platelet NBEAL2 has been reported for others. GPS is commonly associated with mild bleeding, but lifethreatening bleeding has been reported in some cases. A common long-term complication in GPS patients is myelofibrosis; splenomegaly is less common but sometimes of sufficient severity to merit splenectomy. Like GPS patients, mice lacking NBEAL2 expression exhibit macrothrombocytopenia, deficiency of platelet α-granules, splenomegaly, myelofibrosis, impaired platelet function and abnormalities in megakaryocyte development. Animal studies have also reported impaired platelet function in vivo using laser injury and thrombo-inflammation models. NBEAL2 is a large gene with 54 exons, and several putative functional domains have been identified in NBEAL2, including PH (pleckstrin homology) and BEACH (beige and Chediak-Higashi) domains shared with other members of a protein family that includes LYST and LRBA, also expressed by hematopoietic cells. Potential NBEAL2-interacting proteins have recently been identified, and it is expected that current and future efforts will reveal the cellular mechanisms by which NBEAL2 facilitates platelet development and supports hemostatic function.


Assuntos
Plaquetas/metabolismo , Proteínas Sanguíneas/genética , Síndrome da Plaqueta Cinza/genética , Mutação , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Sanguíneas/metabolismo , Criança , Pré-Escolar , Feminino , Síndrome da Plaqueta Cinza/sangue , Síndrome da Plaqueta Cinza/metabolismo , Hemorragia/sangue , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Adulto Jovem
6.
Platelets ; 28(2): 147-154, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28277061

RESUMO

Platelets are critical to hemostasis and thrombosis. Upon detecting injury, platelets show a range of responses including the release of protein cargo from α-granules. This cargo is synthesized by platelet precursor megakaryocytes or endocytosed by megakaryocytes and/or platelets. Insights into α-granule biogenesis have come from studies of hereditary conditions where these granules are immature, deficient or absent. Studies of Arthrogryposis, Renal dysfunction, and Cholestasis (ARC) syndrome identified the first proteins essential to α-granule biogenesis: VPS33B and VPS16B. VPS33B and VPS16B form a complex, and in the absence of either, platelets lack α-granules and the granule-specific membrane protein P-selectin. Gray Platelet Syndrome (GPS) platelets also lack conventionally recognizable α-granules, although P-selectin containing structures are present. GPS arises from mutations affecting NBEAL2. The GPS phenotype is more benign than ARC syndrome, but it can cause life-threatening bleeding, progressive thrombocytopenia, and myelofibrosis. We review the essential roles of VPS33B, VPS16B, and NBEAL2 in α-granule development. We also examine the existing data on their mechanisms of action, where many details remain poorly understood. VPS33B and VPS16B are ubiquitously expressed and ARC syndrome is a multisystem disorder that causes lethality early in life. Thus, VPS33B and VPS16B are clearly involved in other processes besides α-granule biogenesis. Studies of their involvement in vesicular trafficking and protein interactions are reviewed to gain insights into their roles in α-granule formation. NBEAL2 mutations primarily affect megakaryocytes and platelets, and while little is known about NBEAL2 function some insights can be gained from studies of related proteins, such as LYST.


Assuntos
Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Animais , Artrogripose/diagnóstico , Artrogripose/etiologia , Artrogripose/metabolismo , Transporte Biológico , Plaquetas/ultraestrutura , Colestase/diagnóstico , Colestase/etiologia , Colestase/metabolismo , Grânulos Citoplasmáticos/ultraestrutura , Síndrome da Plaqueta Cinza/diagnóstico , Síndrome da Plaqueta Cinza/etiologia , Síndrome da Plaqueta Cinza/metabolismo , Humanos , Megacariócitos/metabolismo , Mutação , Fenótipo , Insuficiência Renal/diagnóstico , Insuficiência Renal/etiologia , Insuficiência Renal/metabolismo , Vesículas Secretórias/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
7.
Sci Rep ; 6: 23213, 2016 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-26987485

RESUMO

The Gray Platelet Syndrome (GPS) is a rare inherited bleeding disorder characterized by deficiency of platelet α-granules, macrothrombocytopenia and marrow fibrosis. The autosomal recessive form of GPS is linked to loss of function mutations in NBEAL2, which is predicted to regulate granule trafficking in megakaryocytes, the platelet progenitors. We report the first analysis of cultured megakaryocytes from GPS patients with NBEAL2 mutations. Megakaryocytes cultured from peripheral blood or bone marrow hematopoietic progenitor cells from four patients were used to investigate megakaryopoiesis, megakaryocyte morphology and platelet formation. In vitro differentiation of megakaryocytes was normal, whereas we observed deficiency of megakaryocyte α-granule proteins and emperipolesis. Importantly, we first demonstrated that platelet formation by GPS megakaryocytes was severely affected, a defect which might be the major cause of thrombocytopenia in patients. These results demonstrate that cultured megakaryocytes from GPS patients provide a valuable model to understand the pathogenesis of GPS in humans.


Assuntos
Plaquetas/citologia , Proteínas Sanguíneas/genética , Síndrome da Plaqueta Cinza/patologia , Megacariócitos/citologia , Idoso , Plaquetas/metabolismo , Cálcio/metabolismo , Diferenciação Celular , Células Cultivadas , Criança , Emperipolese , Síndrome da Plaqueta Cinza/genética , Síndrome da Plaqueta Cinza/metabolismo , Humanos , Masculino , Megacariócitos/metabolismo , Megacariócitos/patologia , Modelos Biológicos , Mutação , Adulto Jovem
9.
Platelets ; 26(8): 751-7, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25806575

RESUMO

The gray platelet syndrome (GPS) is a rare congenital platelet disorder characterized by mild to moderate bleeding diathesis, macrothrombocytopenia and lack of azurophilic α-granules in platelets. Some platelet and megakaryocyte (MK) abnormalities have been described, but confirmative studies of the defects in larger patient cohorts have not been undertaken. We studied platelet function and bone marrow (BM) features in five GPS patients with NBEAL2 autosomal recessive mutations from four unrelated families. In 3/3 patients, we observed a defect in platelet responses to protease-activated receptor (PAR)1-activating peptide as the most consistent finding, either isolated or combined to defective responses to other agonists. A reduction of PAR1 receptors with normal expression of major glycoproteins on the platelet surface was also found. Thrombin-induced fibrinogen binding to platelets was severely impaired in 2/2 patients. In 4/4 patients, the BM biopsy showed fibrosis (grade 2-3) and extensive emperipolesis, with many (36-65%) MKs containing 2-4 leukocytes engulfed within the cytoplasm. Reduced immunolabeling for platelet factor 4 together with normal immunolabeling for CD63 in MKs of two patients demonstrated that GPS MKs display an alpha granule-specific defect. Increased immunolabeling for P-selectin and decreased immunolabeling for PAR1, PAR4 and c-MPL were also observed in MKs of two patients. Marked emperipolesis, specific defect of MK alpha-granule content and defect of PAR1-mediated platelet responses are present in all GPS patients that we could study in detail. These results help to further characterize the disease.


Assuntos
Plaquetas/metabolismo , Emperipolese , Síndrome da Plaqueta Cinza/metabolismo , Megacariócitos/metabolismo , Trifosfato de Adenosina/metabolismo , Idoso , Plaquetas/patologia , Proteínas Sanguíneas/genética , Medula Óssea/metabolismo , Medula Óssea/patologia , Estudos de Casos e Controles , Grânulos Citoplasmáticos/metabolismo , Grânulos Citoplasmáticos/patologia , Fibrose , Expressão Gênica , Síndrome da Plaqueta Cinza/diagnóstico , Síndrome da Plaqueta Cinza/genética , Humanos , Megacariócitos/patologia , Mutação , Ativação Plaquetária , Agregação Plaquetária , Testes de Função Plaquetária , Receptor PAR-1/genética , Receptor PAR-1/metabolismo
10.
Blood ; 124(24): 3624-35, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25258341

RESUMO

NBEAL2 encodes a multidomain scaffolding protein with a putative role in granule ontogeny in human platelets. Mutations in NBEAL2 underlie gray platelet syndrome (GPS), a rare inherited bleeding disorder characterized by a lack of α-granules within blood platelets and progressive bone marrow fibrosis. We present here a novel Nbeal2(-/-) murine model of GPS and demonstrate that the lack of α-granules is due to their loss from platelets/mature megakaryocytes (MKs), and not by initial impaired formation. We show that the lack of Nbeal2 confers a proinflammatory phenotype to the bone marrow MKs, which in combination with the loss of proteins from α-granules drives the development of bone marrow fibrosis. In addition, we demonstrate that α-granule deficiency impairs platelet function beyond their purely hemostatic role and that Nbeal2 deficiency has a protective effect against cancer metastasis.


Assuntos
Síndrome da Plaqueta Cinza/metabolismo , Megacariócitos/metabolismo , Animais , Proteínas Sanguíneas/genética , Proteínas Sanguíneas/metabolismo , Modelos Animais de Doenças , Síndrome da Plaqueta Cinza/genética , Síndrome da Plaqueta Cinza/patologia , Humanos , Megacariócitos/patologia , Camundongos , Camundongos Knockout , Mutação , Metástase Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Mielofibrose Primária/genética , Mielofibrose Primária/metabolismo , Mielofibrose Primária/patologia , Vesículas Secretórias
11.
Blood ; 122(19): 3349-58, 2013 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-23861251

RESUMO

Gray platelet syndrome (GPS) is an inherited bleeding disorder associated with macrothrombocytopenia and α-granule-deficient platelets. GPS has been linked to loss of function mutations in NEABL2 (neurobeachin-like 2), and we describe here a murine GPS model, the Nbeal2(-/-) mouse. As in GPS, Nbeal2(-/-) mice exhibit splenomegaly, macrothrombocytopenia, and a deficiency of platelet α-granules and their cargo, including von Willebrand factor (VWF), thrombospondin-1, and platelet factor 4. The platelet α-granule membrane protein P-selectin is expressed at 48% of wild-type levels and externalized upon platelet activation. The presence of P-selectin and normal levels of VPS33B and VPS16B in Nbeal2(-/-) platelets suggests that NBEAL2 acts independently of VPS33B/VPS16B at a later stage of α-granule biogenesis. Impaired Nbeal2(-/-) platelet function was shown by flow cytometry, platelet aggregometry, bleeding assays, and intravital imaging of laser-induced arterial thrombus formation. Microscopic analysis detected marked abnormalities in Nbeal2(-/-) bone marrow megakaryocytes, which when cultured showed delayed maturation, decreased survival, decreased ploidy, and developmental abnormalities, including abnormal extracellular distribution of VWF. Our results confirm that α-granule secretion plays a significant role in platelet function, and they also indicate that abnormal α-granule formation in Nbeal2(-/-) mice has deleterious effects on megakaryocyte survival, development, and platelet production.


Assuntos
Plaquetas/patologia , Proteínas Sanguíneas/genética , Medula Óssea/patologia , Grânulos Citoplasmáticos/patologia , Síndrome da Plaqueta Cinza/patologia , Megacariócitos/patologia , Animais , Plaquetas/metabolismo , Proteínas Sanguíneas/deficiência , Medula Óssea/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diferenciação Celular , Grânulos Citoplasmáticos/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Síndrome da Plaqueta Cinza/genética , Síndrome da Plaqueta Cinza/metabolismo , Masculino , Megacariócitos/metabolismo , Camundongos , Camundongos Knockout , Selectina-P/genética , Selectina-P/metabolismo , Agregação Plaquetária , Fator Plaquetário 4/genética , Fator Plaquetário 4/metabolismo , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Trombospondina 1/genética , Trombospondina 1/metabolismo , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA