Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 181
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 132(6): 622-634, 2018 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-29794068

RESUMO

Thrombopoietin (TPO), a hematopoietic growth factor produced predominantly by the liver, is essential for thrombopoiesis. Prevailing theory posits that circulating TPO levels are maintained through its clearance by platelets and megakaryocytes via surface c-Mpl receptor internalization. Interestingly, we found a two- to threefold decrease in circulating TPO in GPIbα-/- mice compared with wild-type (WT) controls, which was consistent in GPIbα-deficient human Bernard-Soulier syndrome (BSS) patients. We showed that lower TPO levels in GPIbα-deficient conditions were not due to increased TPO clearance by GPIbα-/- platelets but rather to decreased hepatic TPO mRNA transcription and production. We found that WT, but not GPIbα-/-, platelet transfusions rescued hepatic TPO mRNA and circulating TPO levels in GPIbα-/- mice. In vitro hepatocyte cocultures with platelets or GPIbα-coupled beads further confirm the disruption of platelet-mediated hepatic TPO generation in the absence of GPIbα. Treatment of GPIbα-/- platelets with neuraminidase caused significant desialylation; however, strikingly, desialylated GPIbα-/- platelets could not rescue impaired hepatic TPO production in vivo or in vitro, suggesting that GPIbα, independent of platelet desialylation, is a prerequisite for hepatic TPO generation. Additionally, impaired hepatic TPO production was recapitulated in interleukin-4/GPIbα-transgenic mice, as well as with antibodies targeting the extracellular portion of GPIbα, demonstrating that the N terminus of GPIbα is required for platelet-mediated hepatic TPO generation. These findings reveal a novel nonredundant regulatory role for platelets in hepatic TPO homeostasis, which improves our understanding of constitutive TPO regulation and has important implications in diseases related to GPIbα, such as BSS and auto- and alloimmune-mediated thrombocytopenias.


Assuntos
Síndrome de Bernard-Soulier/sangue , Plaquetas/fisiologia , Fígado/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Trombopoetina/biossíntese , Animais , Síndrome de Bernard-Soulier/genética , Células Cultivadas , Glicosilação , Hepatócitos/metabolismo , Homeostase , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Ácido N-Acetilneuramínico/metabolismo , Transfusão de Plaquetas , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes/metabolismo , Trombopoetina/sangue
2.
Microvasc Res ; 132: 104039, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32645366

RESUMO

Implantable medical devices and biosensors are pivotal in revolutionizing the field of medical technology by opening new dimensions in the field of disease detection and cure. These devices need to harness a biocompatible and physiologically sustainable safe power source instead of relying on external stimuli, overcoming the constraints on their applicability in-vivo. Here, by appealing to the interplay of electromechanics and hydrodynamics in physiologically relevant microvessels, we bring out the role of charged endothelial glycocalyx layer (EGL) towards establishing a streaming potential across physiological fluidic conduits. We account for the complex rheology of blood-mimicking fluid by appealing to Newtonian fluid model representing the blood plasma and a viscoelastic fluid model representing the whole blood. We model the EGL as a poroelastic layer with volumetric charge distribution. Our results reveal that for physiologically relevant micro-flows, the streaming potential induced is typically of the order of 0.1 V/mm, which may turn out to be substantial towards energizing biosensors and implantable medical devices whose power requirements are typically in the range of micro to milliwatts. We also bring out the specific implications of the relevant physiological parameters towards establishment of the streaming potential, with a vision of augmenting the same within plausible functional limits. We further unveil that the dependence of streaming potential on EGL thickness might be one of the key aspects in unlocking the mystery behind the angiogenesis pattern. Our results may open up novel bio-sensing and actuating possibilities in medical diagnostics as well as may provide a possible alternative regarding the development of physiologically safe and biocompatible power sources within the human body.


Assuntos
Materiais Biomiméticos , Capilares/fisiologia , Células Endoteliais/fisiologia , Microcirculação , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Substâncias Viscoelásticas/química , Velocidade do Fluxo Sanguíneo , Hemorreologia , Humanos , Hidrodinâmica , Modelos Cardiovasculares
3.
Hepatology ; 62(5): 1593-605, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26202541

RESUMO

UNLABELLED: Although platelets have been extensively studied in hemostasis and inflammation, their role is not well understood in sterile liver injury and repair. Using a thermally induced focal liver injury and repair model and multichannel spinning disk confocal microscopy allowed visualization of the dynamic behavior of platelets and neutrophils in this insult. Platelets instantaneously adhered to molecularly altered sinusoidal endothelium adjacent to the afflicted area, paving approximately 200 µm abutting the injury. Platelets remained adherent for at least 4 hours, but dissipated by 8 hours. The early recruitment occurred by GPIIbIIIa (CD41) and the later recruitment was dependent upon both GPIIbIIIa and GPIb (CD42B). Platelets did not occlude the vessels, but rather paved the altered endothelium. Endothelin-induced vasoconstriction by hepatic stellate cells, and not platelet accumulation or coagulation, was responsible for temporarily restricted perfusion around the injury. Neutrophils crawled into the injury from significant distances through the sinusoids. The crawling neutrophils required the platelet-paved endothelium given that very little neutrophil recruitment was noted in thrombocytopenic or CD41-deficient mice. As platelets slowly dissipated, neutrophil recruitment was also halted. Previous work suggested that platelets binding to immobilized neutrophils induced neutrophil extracellular trap (NET) formation in response to infection as well as during thrombosis and other forms of sterile injury. In this model of neutrophils crawling on immobilized platelets, very few NETs were observed and no additional injury was noted. In fact, GPIIbIIIa-deficient mice had delayed repair. CONCLUSION: In a liver model of sterile injury and repair, platelets play a critical role in forming a substratum and pave the way for neutrophils to enter the injured site for subsequent repair.


Assuntos
Plaquetas/fisiologia , Comunicação Celular , Fígado/lesões , Neutrófilos/fisiologia , Animais , Endotélio Vascular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Glicoproteína IIb da Membrana de Plaquetas/fisiologia
4.
Ann Neurol ; 75(5): 670-83, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24644058

RESUMO

OBJECTIVE: Bacterial infection contributes to diverse noninfectious diseases and worsens outcome after stroke. Streptococcus pneumoniae, the most common infection in patients at risk of stroke, is a major cause of prolonged hospitalization and death of stroke patients, but how infection impacts clinical outcome is not known. METHODS: We induced sustained pulmonary infection by a human S. pneumoniae isolate in naive and comorbid rodents to investigate the effect of infection on vascular and inflammatory responses prior to and after cerebral ischemia. RESULTS: S. pneumoniae infection triggered atherogenesis, led to systemic induction of interleukin (IL) 1, and profoundly exacerbated (50-90%) ischemic brain injury in rats and mice, a response that was more severe in combination with old age and atherosclerosis. Systemic blockade of IL-1 with IL-1 receptor antagonist (IL-1Ra) fully reversed infection-induced exacerbation of brain injury and functional impairment caused by cerebral ischemia. We show that infection-induced systemic inflammation mediates its effects via increasing platelet activation and microvascular coagulation in the brain after cerebral ischemia, as confirmed by reduced brain injury in response to blockade of platelet glycoprotein (GP) Ibα. IL-1 and platelet-mediated signals converge on microglia, as both IL-1Ra and GPIbα blockade reversed the production of IL-1α by microglia in response to cerebral ischemia in infected animals. INTERPRETATION: S. pneumoniae infection augments atherosclerosis and exacerbates ischemic brain injury via IL-1 and platelet-mediated systemic inflammation. These mechanisms may contribute to diverse cardio- and cerebrovascular pathologies in humans.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Interleucina-1/efeitos adversos , Complexo Glicoproteico GPIb-IX de Plaquetas/efeitos adversos , Infecções Estreptocócicas/metabolismo , Infecções Estreptocócicas/patologia , Streptococcus pneumoniae , Animais , Isquemia Encefálica/microbiologia , Progressão da Doença , Humanos , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Interleucina-1/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/metabolismo , Microglia/microbiologia , Microglia/patologia , Ativação Plaquetária , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Ratos , Ratos Wistar , Infecções Estreptocócicas/microbiologia
5.
Blood ; 122(6): 1026-33, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23801636

RESUMO

Previously we reported that dietary intake of alpha-linolenic acid (ALA) reduces atherogenesis and inhibits arterial thrombosis. Here, we analyze the substantial increase in platelet count induced by ALA and the mechanisms of reduced platelet clearance. Eight-week-old male apolipoprotein E knockout (ApoE(-/-)) mice were fed a 0.21g% cholesterol diet complemented by either a high- (7.3g%) or low-ALA (0.03g%) content. Platelet counts doubled after 16 weeks of ALA feeding, whereas the bleeding time remained similar. Plasma glycocalicin and glycocalicin index were reduced, while reticulated platelets, thrombopoietin, and bone marrow megakaryocyte colony-forming units remained unchanged. Platelet contents of liver and spleen were substantially reduced, without affecting macrophage function and number. Glycoprotein Ib (GPIb) shedding, exposure of P-selectin, and activated integrin αIIbß3 upon activation with thrombin were reduced. Dietary ALA increased the platelet count by reducing platelet clearance in the reticulo-endothelial system. The latter appears to be mediated by reduced cleavage of GPIb by tumor necrosis factor-α-converting enzyme and reduced platelet activation/expression of procoagulant signaling. Ex vivo, there was less adhesion of human platelets to von Willebrand factor under high shear conditions after ALA treatment. Thus, ALA may be a promising tool in transfusion medicine and in high turnover/high activation platelet disorders.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Plaquetas/efeitos dos fármacos , Ácido alfa-Linolênico/uso terapêutico , Proteínas ADAM/metabolismo , Proteína ADAM17 , Ração Animal , Animais , Aterosclerose/metabolismo , Plaquetas/metabolismo , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Selectina-P/metabolismo , Ativação Plaquetária , Adesividade Plaquetária , Contagem de Plaquetas , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Transdução de Sinais , Baço/metabolismo
6.
Adv Exp Med Biol ; 844: 263-75, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25480646

RESUMO

At the clinical level, recent studies reveal the link between coagulation and other pathophysiological processes, including platelet activation, inflammation, cancer, the immune response, and/or infectious diseases. These links are likely to underpin the coagulopathy associated with risk factors for venous thromboembolic (VTE) and deep vein thrombosis (DVT). At the molecular level, the interactions between platelet-specific receptors and coagulation factors could help explain coagulopathy associated with aberrant platelet function, as well as revealing new approaches targeting platelet receptors in diagnosis or treatment of VTE or DVT. Glycoprotein (GP)Ibα, the major ligand-binding subunit of the platelet GPIb-IX-V complex, that binds the adhesive ligand, von Willebrand factor (VWF), is co-associated with the platelet-specific collagen receptor, GPVI. The GPIb-IX-V/GPVI adheso-signaling complex not only initiates platelet activation and aggregation (thrombus formation) in response to vascular injury or disease but GPIbα also regulates coagulation through a specific interaction with thrombin and other coagulation factors. Here, we discuss the structure and function of key platelet receptors involved in thrombus formation and coagulation in health and disease, with a particular focus on platelet GPIbα.


Assuntos
Coagulação Sanguínea , Complexo Glicoproteico GPIb-IX de Plaquetas/química , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Glicoproteínas da Membrana de Plaquetas/química , Glicoproteínas da Membrana de Plaquetas/fisiologia , Animais , Plaquetas/fisiologia , Humanos , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade
7.
Blood ; 117(18): 4999-5008, 2011 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-21398580

RESUMO

The membrane-anchored CX3C chemokine fractalkine (FKN) is expressed on activated endothelium and is associated with the development of atherosclerosis. The potential of FKN in mediating platelet adhesion beyond platelet activation remains unexplored to date. A flow-based adhesion assay was used to study the adhesion of platelets to immobilized FKN under physiologic flow conditions. Platelet adhesion to von Willebrand factor (VWF) was increased in the presence of FKN at 600 inverse seconds. Additional platelet adhesion to FKN coimmobilized with VWF was dependent on the FKN receptor CX3CR1 and activation of glycoprotein (GP) IIb/IIIa. The number of platelets rolling on VWF was likewise enhanced in the presence of FKN. The enhancement of rolling on FKN and VWF was insensitive to anti-CX3CR1 antibody but was fully inhibited by neutralizing GPIbα function. The extracellular domain of GPIbα was covalently coupled to fluorescent microspheres, and microsphere binding was significantly higher in the presence of FKN. Platelet adhesion to activated endothelium in vitro and to intact human arteries was substantially increased in an FKN-dependent manner. These data demonstrate that endothelial expressed FKN activates platelets via its cognate receptor CX3CR1, whereas platelet adhesion is predominantly mediated by GPIbα and independent of CX3CR1.


Assuntos
Quimiocina CX3CL1/fisiologia , Adesividade Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Glicoproteínas da Membrana de Plaquetas/fisiologia , Fator de von Willebrand/fisiologia , Artérias/fisiologia , Receptor 1 de Quimiocina CX3C , Células Endoteliais/fisiologia , Hemorreologia , Humanos , Técnicas In Vitro , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Receptores de Citocinas/antagonistas & inibidores , Receptores de Citocinas/fisiologia , Receptores de HIV/antagonistas & inibidores , Receptores de HIV/fisiologia
8.
Blood ; 116(7): 1157-64, 2010 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-20457869

RESUMO

Activated platelets become procoagulant and efficiently promote the conversion of prothrombin to thrombin. A role of the GPIb-V-IX complex has long been postulated in view of the decreased prothrombin consumption in Bernard-Soulier patients. We evaluated the impact of GPIb-V-IX deficiency and the requirement for the GPIbalpha extracellular domain. In GPIbbeta(-/-) mice, thrombin generation was profoundly decreased in tissue factor- or collagen-related peptide (CRP)-activated platelet-rich plasma and in washed platelets supplemented with normal plasma or with FVa, FXa, and prothrombin. Phosphatidylserine (PS) exposure was similarly decreased in response to thrombin, CRP, or CRP + PAR4 peptide despite a normal platelet phospholipid composition. The hypothesis that these defects originate from lack of the GPIbalpha N-terminal domain was evaluated after its removal from normal mouse and human platelets with Nk protease or O-sialoglycoprotein endopeptidase. Unexpectedly, the treated platelets exhibited normal thrombin generation and PS exposure, indicating that GPIb-V-IX regulates procoagulant activity independently of its GPIbalpha-binding region. These results suggested a more general structuring role through intracellular cytoskeleton-anchoring portions regulating responses leading to PS exposure. This hypothesis was supported by the decreased calcium mobilization observed in GPIbbeta(-/-) platelets in response to several agonists, some acting independently of GPIb, in contrast to the normal calcium responses in Nk protease-treated platelets.


Assuntos
Plaquetas/metabolismo , Glicoproteínas de Membrana/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Animais , Cálcio/metabolismo , Citometria de Fluxo , Granzimas/metabolismo , Humanos , Camundongos , Camundongos Knockout , Fosfatidilserinas/metabolismo , Estrutura Terciária de Proteína , Trombina/metabolismo
9.
Handb Exp Pharmacol ; (210): 287-309, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22918736

RESUMO

The formation of platelet-rich thrombi, a critical step in the pathogenesis of atherothrombotic events, is a multistep process involving several components, among which von Willebrand Factor (VWF) plays a central role. Ruptured atherosclerotic plaques expose subendothelial matrix proteins which bind VWF that represents a bridge between the injured blood vessel and activated platelets, playing a crucial role in platelet adhesion and aggregation, especially in conditions of high-shear rate. Due to these peculiarities, the binding of VWF to GPIbα is an attractive drug target. Here we summarize the present knowledge on the different classes of drugs targeting the VWF-GPIb interaction and we give an account of their level of clinical development. In particular, the following compounds are discussed: AJW200, an IgG4 humanized monoclonal antibody against VWF-A1; 82D6A3, a monoclonal antibody against VWF-A3; ALX-0081 and ALX-0681, bivalent humanized nanobodies targeting the VWF-A1 domain; ARC1779 and its advanced formulation ARC15105, second-generation aptamers that bind the VWF-A1 domain; h6B4-Fab, a murine monoclonal antibody, and GPG-290, a recombinant chimeric protein, both directed against GPIbα.


Assuntos
Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Fator de von Willebrand/antagonistas & inibidores , Animais , Aptâmeros de Nucleotídeos/uso terapêutico , Humanos , Ativação Plaquetária , Inibidores da Agregação Plaquetária/uso terapêutico , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Proteínas Recombinantes de Fusão/farmacologia , Proteínas Recombinantes de Fusão/uso terapêutico , Trombose/tratamento farmacológico , Fator de von Willebrand/química , Fator de von Willebrand/fisiologia
10.
J Exp Med ; 192(2): 193-204, 2000 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-10899906

RESUMO

The firm adhesion and transplatelet migration of leukocytes on vascular thrombus are both dependent on the interaction of the leukocyte integrin, Mac-1, and a heretofore unknown platelet counterreceptor. Here, we identify the platelet counterreceptor as glycoprotein (GP) Ibalpha, a component of the GP Ib-IX-V complex, the platelet von Willebrand factor (vWf) receptor. THP-1 monocytic cells and transfected cells that express Mac-1 adhered to GP Ibalpha-coated wells. Inhibition studies with monoclonal antibodies or receptor ligands showed that the interaction involves the Mac-1 I domain (homologous to the vWf A1 domain), and the GP Ibalpha leucine-rich repeat and COOH-terminal flanking regions. The specificity of the interaction was confirmed by the finding that neutrophils from wild-type mice, but not from Mac-1-deficient mice, bound to purified GP Ibalpha and to adherent platelets, the latter adhesion being inhibited by pretreatment of the platelets with mocarhagin, a protease that specifically cleaves GP Ibalpha. Finally, immobilized GP Ibalpha supported the rolling and firm adhesion of THP-1 cells under conditions of flow. These observations provide a molecular target for disrupting leukocyte-platelet complexes that promote vascular inflammation in thrombosis, atherosclerosis, and angioplasty-related restenosis.


Assuntos
Antígeno de Macrófago 1/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Animais , Sítios de Ligação , Plaquetas/fisiologia , Adesão Celular , Linhagem Celular , Humanos , Antígeno-1 Associado à Função Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/fisiologia
11.
Hamostaseologie ; 30(3): 136-8, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20680228

RESUMO

Ischaemic stroke is a devastating disease with limited treatment options due to numerous uncertainties regarding the underlying pathophysiology. The contribution of glycoprotein (GP)Ibalpha and von Willebrand factor (VWF) in stroke development has only recently been established in mice. Complete blockade of GPIbalpha led to a significant reduction of infarct volumes in mice undergoing one hour of transient middle cerebral artery occlusion (tMCAO). High shear-induced changes in VWF confirmation are a prerequisite for VWF binding to collagen and GPIbalpha expressed on platelets. Importantly, transgenic VWF-/- mice were similarly protected against ischemic stroke after tMCAO, and hydrodynamic injection of a VWF-encoding plasmid restored VWF serum levels and the susceptibility towards stroke. Secreted VWF is rapidly cleaved by ADAMTS13. Accordingly, ADAMTS13 deficient mice developed larger infarction after tMCAO, while infusion of recombinant ADAMTS13 into wild-type mice was stroke-protective. In conclusion, there is compelling evidence that GPIbalpha/VWF interactions and downstream signaling via phospholipase D1 (PLD1) provide new therapeutic targets in ischemic stroke.


Assuntos
Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Acidente Vascular Cerebral/sangue , Fator de von Willebrand/fisiologia , Animais , Humanos , Camundongos , Acidente Vascular Cerebral/fisiopatologia , Acidente Vascular Cerebral/prevenção & controle
12.
J Cell Biol ; 147(5): 1085-96, 1999 Nov 29.
Artigo em Inglês | MEDLINE | ID: mdl-10579727

RESUMO

We have reconstituted the platelet glycoprotein (GP) Ib-IX-mediated activation of the integrin alpha(IIb)beta(3) in a recombinant DNA expression model, and show that 14-3-3 is important in GPIb-IX signaling. CHO cells expressing alpha(IIb)beta(3) adhere poorly to vWF. Cells expressing GPIb-IX adhere to vWF in the presence of botrocetin but spread poorly. Cells coexpressing integrin alpha(IIb)beta(3) and GPIb-IX adhere and spread on vWF, which is inhibited by RGDS peptides and antibodies against alpha(IIb)beta(3). vWF binding to GPIb-IX also activates soluble fibrinogen binding to alpha(IIb)beta(3) indicating that GPIb-IX mediates a cellular signal leading to alpha(IIb)beta(3) activation. Deletion of the 14-3-3-binding site in GPIbalpha inhibited GPIb-IX-mediated fibrinogen binding to alpha(IIb)beta(3) and cell spreading on vWF. Thus, 14-3-3 binding to GPIb-IX is important in GPIb-IX signaling. Expression of a dominant negative 14-3-3 mutant inhibited cell spreading on vWF, suggesting an important role for 14-3-3. Deleting both the 14-3-3 and filamin-binding sites of GPIbalpha induced an endogenous integrin-dependent cell spreading on vWF without requiring alpha(IIb)beta(3), but inhibited vWF-induced fibrinogen binding to alpha(IIb)beta(3). Thus, while different activation mechanisms may be responsible for vWF interaction with different integrins, GPIb-IX-mediated activation of alpha(IIb)beta(3) requires 14-3-3 interaction with GPIbalpha.


Assuntos
Regulação da Expressão Gênica , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/biossíntese , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Proteínas/fisiologia , Transfecção , Tirosina 3-Mono-Oxigenase , Proteínas 14-3-3 , Sequência de Aminoácidos/genética , Animais , Sítios de Ligação/genética , Células CHO , Proteínas de Transporte/metabolismo , Adesão Celular/genética , Adesão Celular/fisiologia , Proteínas Contráteis/metabolismo , Cricetinae , Citoplasma/química , Fibrinogênio/metabolismo , Filaminas , Proteínas dos Microfilamentos/metabolismo , Peptídeos/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Deleção de Sequência/genética , Transdução de Sinais , Fator de von Willebrand/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 28(2): 329-34, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17962622

RESUMO

OBJECTIVE: Thrombin containing the mutations Trp215Ala and Glu217Ala (WE) selectively activates protein C and has potent antithrombotic effects in primates. The aim of this study was to delineate the molecular mechanism of direct WE-platelet interactions under static and shear conditions. METHODS AND RESULTS: Purified platelets under static conditions bound and spread on immobilized wild-type but not WE thrombin. In PPACK-anticoagulated blood under shear flow conditions, platelets tethered and rolled on both wild-type and WE thrombin, and these interactions were abrogated by the presence of a glycoprotein Ib (GPIb)-blocking antibody. Platelet deposition on collagen was blocked in the presence of WE, but not wild-type thrombin or prothrombin. WE also abrogated platelet tethering and rolling on immobilized von Willebrand factor in whole blood under shear flow. CONCLUSIONS: These observations demonstrate that the thrombin mutant WE, while not activating platelets, retains the ability to interact with platelets through GPIb, and inhibits GPIb-dependent binding to von Willebrand factor-collagen under shear.


Assuntos
Coagulação Sanguínea/fisiologia , Proteínas Mutantes/fisiologia , Ativação Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Trombina/genética , Trombina/fisiologia , Adulto , Animais , Plaquetas/fisiologia , Enzimas Imobilizadas , Humanos , Camundongos , Adesividade Plaquetária/fisiologia , Proteínas Recombinantes , Reologia , Trombina/química , Trombose/fisiopatologia , Fator de von Willebrand/fisiologia
14.
Placenta ; 29(9): 826-32, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18684502

RESUMO

Microvesicles (MVs) can derive from several cell types and their membranes contain cell surface elements. Their role is increasingly recognized in cell-to-cell communication, as they act as both paracrine and remote messengers, occurring in circulating form as well as in plasma. Successful pregnancy requires a series of interactions between the maternal immune system and the implanted fetus, such that the semi-allograft will not be rejected. These interactions occur at the materno-placental interface and/or at a systemic level. In the present study we identified for the first time the in vivo plasma pattern of the MVs of third-trimester, healthy pregnant women, their cellular origin, and their target cells using flow cytometry and confocal laser microscopy. We searched for the cellular target molecules of thrombocyte-derived MVs with the help of neutralizing antibodies. We examined the in vitro effects of MVs on STAT3 phosphorylation of primary lymphocytes and Jurkat cells. We found that both placental trophoblast-derived and maternal thrombocyte-derived MVs bind to circulating peripheral T lymphocytes, but not to B lymphocytes or NK cells. We were able to show that the P-selectin (CD62P)-PSGL-1 (CD162) interaction is one mechanism binding platelet-derived MVs to T cells. We were also able to demonstrate that MV-lymphocyte interactions induce STAT3 phosphorylation in T cells. Our findings indicate that both thrombocyte- and trophoblast-derived MVs may play an important role in the immunomodulation of pregnancy. We suggest that the transfer of different signals via MVs represents a novel form of communication between the placenta and the maternal immune system, and that MVs contribute to the establishment of stable immune tolerance to the semi-allograft fetus.


Assuntos
Plaquetas/fisiologia , Gravidez/imunologia , Linfócitos T/fisiologia , Vesículas Transportadoras/fisiologia , Trofoblastos/fisiologia , Diferenciação Celular , Feminino , Humanos , Selectina-P/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Vesículas Transportadoras/imunologia
15.
Circ Res ; 98(1): 98-104, 2006 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-16322482

RESUMO

Although platelets do not ordinarily bind to endothelial cells (EC), pathological interactions between platelets and arterial EC may contribute to the propagation of atheroma. Previously, in an in vitro model of atherogenesis, where leukocyte adhesion to EC cocultured with smooth muscle cells was greatly enhanced, we also observed attachment of platelets to the EC layer. Developing this system to specifically model platelet adhesion, we show that EC cocultured with smooth muscle cells can bind platelets in a process that is dependent on EC activation by tumor necrosis factor (TNF)-alpha and transforming growth factor (TGF)-beta1. Recapitulating the model using EC alone, we found that a combination of TGF-beta1 and TNF-alpha promoted high levels of platelet adhesion compared with either agent used in isolation. Platelet adhesion was inhibited by antibodies against GPIb-IX-V or alpha(IIb)beta3 integrin, indicating that both receptors are required for stable adhesion. Platelet activation during interaction with the EC was also essential, as treatment with prostacyclin or theophylline abolished stable adhesion. Confocal microscopy of the surface of EC activated with TNF-alpha and TGF-beta1 revealed an extensive matrix of von Willebrand factor that was able to support the adhesion of flowing platelets at wall shear rates below 400 s(-1). Thus, we have demonstrated a novel route of EC activation which is relevant to the atherosclerotic microenvironment. EC activated in this manner would therefore be capable of recruiting platelets in the low-shear environments that commonly exist at points of atheroma formation.


Assuntos
Aterosclerose/etiologia , Células Endoteliais/citologia , Músculo Liso Vascular/citologia , Miócitos de Músculo Liso/fisiologia , Adesividade Plaquetária , Aterosclerose/patologia , Células Cultivadas , Técnicas de Cocultura , Humanos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Fator de Necrose Tumoral alfa/farmacologia , Fator de von Willebrand/fisiologia
16.
Thromb Res ; 121(4): 433-41, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-17644165

RESUMO

Allografts are currently short for clinical potential recipients. Organs and tissues from pigs could be a potential alternative source for clinical transplantation because of their high similarity in anatomical and physiological aspects. Thrombosis could be a consequence of the immunological response or the physiological incompatibilities in cell and molecular levels across species. Platelets play an essential role in haemostasis and the incompatibility of platelets between pigs and humans could be related to rejection and dysfunction of xenografts. Pig blood components, including plasma, leukocytes, red blood cells and platelets, could induce aggregation of human platelets directly, which then resulted in severe thrombosis after xenotransplantation. On the other hand, the existence of potential incompatibilities in coagulation and fibrinolytic system between pigs and humans in the context of xenotransplantation is an important consideration. Here we reviewed platelet incompatibility between pigs and humans related to thrombosis after xenotransplantation, and contribution of immunosuppressive agents to minimizing thrombosis and rejection.


Assuntos
Agregação Plaquetária , Trombose/etiologia , Transplante Heterólogo/imunologia , Animais , Complexo Antígeno-Anticorpo/fisiologia , Fatores de Coagulação Sanguínea/fisiologia , Proteínas do Sistema Complemento/fisiologia , Rejeição de Enxerto , Humanos , Imunossupressores/uso terapêutico , Leucócitos Mononucleares/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Suínos
17.
Biol Psychiatry ; 83(8): 692-706, 2018 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-29305086

RESUMO

BACKGROUND: Alterations in early steps of cortical circuit assembly are thought to play a critical role in vulnerability to schizophrenia (SZ), but the pathogenic impact of SZ-risk mutations on corticogenesis remains to be determined. DiGeorge syndrome critical region 2 (DGCR2) is located in the 22q11.2 locus, whose deletion is a major risk factor for SZ. Moreover, exome sequencing of individuals with idiopathic SZ identified a rare missense mutation in DGCR2, further suggesting that DGCR2 is involved in SZ. METHODS: Here we investigated the function of Dgcr2 and the pathogenic impact of the SZ-risk DGCR2 mutation in mouse corticogenesis using in utero electroporation targeted to projection neurons. RESULTS: Dgcr2 knockdown impaired radial locomotion and final translocation of projection neurons, leading to persistent laminar positioning alterations. The DGCR2 missense SZ-risk mutation had a pathogenic impact on projection neuron laminar allocation by reducing protein expression. Mechanistically, we identified Dgcr2 as a novel member of the Reelin complex, regulating the phosphorylation of Reelin-dependent substrates and the expression of Reelin-dependent transcriptional targets. CONCLUSIONS: Overall, this study provides biological evidence that the SZ-risk gene DGCR2 regulates critical steps of early corticogenesis possibly through a Reelin-dependent mechanism. Additionally, we found that the SZ-risk mutation in DGCR2 has a pathogenic impact on cortical formation by reducing protein expression level, suggesting a functional role for DGCR2 haploinsufficiency in the 22q11.2 deletion syndrome.


Assuntos
Moléculas de Adesão Celular Neuronais/fisiologia , Córtex Cerebral/crescimento & desenvolvimento , Proteínas da Matriz Extracelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurogênese/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Esquizofrenia/genética , Serina Endopeptidases/fisiologia , Animais , Movimento Celular/fisiologia , Eletroporação , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Mutação de Sentido Incorreto , Gravidez , Proteína Reelina , Risco
18.
Sci Rep ; 8(1): 2688, 2018 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-29426910

RESUMO

Since the hemorrhage in severe dengue seems to be primarily related to the defect of the platelet, the possibility that dengue virus (DENV) is selectively tropic for one of its surface receptors was investigated. Flow cytometric data of DENV-infected megakaryocytic cell line superficially expressing human glycoprotein Ib (CD42b) and glycoprotein IIb/IIIa (CD41 and CD41a) were analyzed by our custom-written software in MATLAB. In two-dimensional analyses, intracellular DENV was detected in CD42b+, CD41+ and CD41a+ cells. In three-dimensional analyses, the DENV was exclusively detected in CD42b+ cells but not in CD42b- cells regardless of the other expressions. In single-cell virus-protein analyses, the amount of DENV was directly correlated with those of CD42b at the Pearson correlation coefficient of 0.9. Moreover, RT- PCR and apoptosis assays showed that DENV was able to replicate itself and release its new progeny from the infected CD42b+ cells and eventually killed those cells. These results provide evidence for the involvement of CD42b in DENV infection.


Assuntos
Vírus da Dengue/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Plaquetas/metabolismo , Linhagem Celular , Células Cultivadas , Dengue/virologia , Vírus da Dengue/imunologia , Vírus da Dengue/patogenicidade , Citometria de Fluxo/métodos , Humanos , Megacariócitos/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/fisiologia , Dengue Grave/metabolismo , Tropismo , Tropismo Viral/fisiologia
19.
Curr Biol ; 13(7): R282-4, 2003 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-12676108

RESUMO

One of the impediments precluding long-term storage of platelets for blood transfusion is that refrigerated platelets are rapidly cleared from the circulation upon transfusion. New evidence suggests that this clearance is mediated in the liver by the Mac-1 integrin on Kupffer cells recognizing clustered GPIb receptor on platelets, leading to platelet phagocytosis.


Assuntos
Plaquetas/fisiologia , Antígeno de Macrófago 1/fisiologia , Adesividade Plaquetária/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Glicoproteínas da Membrana de Plaquetas , Transfusão de Sangue , Humanos , Células de Kupffer/fisiologia , Fagocitose/fisiologia , Refrigeração , Temperatura
20.
Curr Biol ; 9(3): R110-2, 1999 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-10021379

RESUMO

Platelet aggregation to form a haemostatic plug, or thrombus, plays a key role in preventing bleeding from a wound. Recent studies have provided new insights into how platelet receptors are deployed during the interactions with the vascular subendothelial matrix that lead to haemostatic plug formation.


Assuntos
Hemostasia/fisiologia , Agregação Plaquetária , Animais , Endotélio Vascular/fisiologia , Proteínas da Matriz Extracelular/fisiologia , Humanos , Integrinas/fisiologia , Oligopeptídeos/fisiologia , Adesividade Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/fisiologia , Complexo Glicoproteico GPIb-IX de Plaquetas/fisiologia , Receptores de Colágeno , Fator de von Willebrand/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa