Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Diabetes ; 65(6): 1714-23, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26936963

RESUMO

Short-term studies in subjects with diabetes receiving glucagon-like peptide 1 (GLP-1)-targeted therapies have suggested a reduced number of cardiovascular events. The mechanisms underlying this unexpectedly rapid effect are not known. We cloned full-length GLP-1 receptor (GLP-1R) mRNA from a human megakaryocyte cell line (MEG-01), and found expression levels of GLP-1Rs in MEG-01 cells to be higher than those in the human lung but lower than in the human pancreas. Incubation with GLP-1 and the GLP-1R agonist exenatide elicited a cAMP response in MEG-01 cells, and exenatide significantly inhibited thrombin-, ADP-, and collagen-induced platelet aggregation. Incubation with exenatide also inhibited thrombus formation under flow conditions in ex vivo perfusion chambers using human and mouse whole blood. In a mouse cremaster artery laser injury model, a single intravenous injection of exenatide inhibited thrombus formation in normoglycemic and hyperglycemic mice in vivo. Thrombus formation was greater in mice transplanted with bone marrow lacking a functional GLP-1R (Glp1r(-/-)), compared with those receiving wild-type bone marrow. Although antithrombotic effects of exenatide were partly lost in mice transplanted with bone marrow from Glp1r(-/-) mice, they were undetectable in mice with a genetic deficiency of endothelial nitric oxide synthase. The inhibition of platelet function and the prevention of thrombus formation by GLP-1R agonists represent potential mechanisms for reduced atherothrombotic events.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Peptídeos/farmacologia , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária/efeitos dos fármacos , Trombose/prevenção & controle , Peçonhas/farmacologia , Animais , Linhagem Celular , Exenatida , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Humanos , Pulmão/metabolismo , Camundongos , Óxido Nítrico Sintase Tipo III/deficiência , Pâncreas/metabolismo
3.
Transfusion ; 55(5): 1040-50, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25403994

RESUMO

BACKGROUND: The evidence supporting plasma transfusion as a means to restore hemostatic control and prevent or treat bleeding is weak, leading to uncertainties as to which proteins affect the therapeutic quality of plasma. Some regulators focus on coagulation Factor (F)VIII activity, but whether this measure reflects overall transfusable plasma efficacy is questionable. We developed a mouse model of coagulopathy in which bleeding outcomes were responsive to plasma transfusion and addressed the relative contributions of FVIII and fibrinogen (Fg) to plasma quality. STUDY DESIGN AND METHODS: Anesthetized mice were rendered coagulopathic by four rounds of exchange of whole blood for washed red blood cells (RBCs) in 5% human albumin solution (HAS), which reduced RBCs, platelets, and plasma protein levels by 55, 66, and 80% of starting levels, in a blood exchange-induced coagulopathy approach (BECA). Before tail vein transection, BECA mice were transfused with HAS, wild-type murine fresh-frozen plasma (WT mFFP), or mFFP from FVIII-/- or Fg-/- knockout mice. BECA mice were also subjected to laser-induced arteriolar injury and thrombus formation quantified by intravital microscopy. RESULTS: Transfusion of WT or FVIII-/- mFFP reduced blood loss by fourfold in BECA mice relative to HAS; Fg-/- mFFP had no effect. WT or FVIII-/- mFFP transfusion, but not that of Fg-/- mFFP, increased thrombus size in laser-injured BECA mice arterioles. Extended refrigerated storage of mFFP did not reduce its antihemorrhagic effects. CONCLUSIONS: The content of Fg, but not FVIII, determined the efficacy of plasma transfusion in coagulopathic mice.


Assuntos
Fator VIII/metabolismo , Fibrinogênio/metabolismo , Hemorragia/terapia , Troca Plasmática/métodos , Animais , Eritrócitos , Feminino , Hemólise , Masculino , Camundongos , Camundongos Knockout
4.
J Mater Chem B ; 3(44): 8623-8628, 2015 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-32262718

RESUMO

Much biomedical equipment consisting of or containing plastic polymer(s) must come into contact with blood - an interaction that, at the molecular level, may unfortunately prompt biological processes with potentially deleterious, short- or long-term effects such as thrombosis. In the present investigation, this problem is alleviated for poly(vinyl chloride) (PVC) through chemical surface modification with an ultrathin, monoethylene glycol-based coating - a transformation that is characterized using X-ray photoelectron spectroscopy (XPS) supplemented by contact angle goniometry (CAG). Antithrombogenic properties are assessed through calculation (for the first 10 min, and after 60 min) of the surface coverage percentage due to platelet adhesion, aggregation and thrombus formation upon continuous exposure to fluorescently-labelled whole human blood. At all shear rates investigated (300, 900, and 1500 s-1), surface coverage decreases by >99% with respect to bare PVC (10 min, short-term contact with blood). Most importantly, antithrombogenic performance is retained for longer-term exposure experiments (60 min), regardless of applied shear rate as well.

5.
J Clin Invest ; 124(10): 4281-93, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25180602

RESUMO

Plasma fibronectin (pFn) has long been suspected to be involved in hemostasis; however, direct evidence has been lacking. Here, we demonstrated that pFn is vital to control bleeding in fibrinogen-deficient mice and in WT mice given anticoagulants. At the site of vessel injury, pFn was rapidly deposited and initiated hemostasis, even before platelet accumulation, which is considered the first wave of hemostasis. This pFn deposition was independent of fibrinogen, von Willebrand factor, ß3 integrin, and platelets. Confocal and scanning electron microscopy revealed pFn integration into fibrin, which increased fibrin fiber diameter and enhanced the mechanical strength of clots, as determined by thromboelastography. Interestingly, pFn promoted platelet aggregation when linked with fibrin but inhibited this process when fibrin was absent. Therefore, pFn may gradually switch from supporting hemostasis to inhibiting thrombosis and vessel occlusion following the fibrin gradient that decreases farther from the injured endothelium. Our data indicate that pFn is a supportive factor in hemostasis, which is vital under both genetic and therapeutic conditions of coagulation deficiency. By interacting with fibrin and platelet ß3 integrin, pFn plays a self-limiting regulatory role in thrombosis, suggesting pFn transfusion may be a potential therapy for bleeding disorders, particularly in association with anticoagulant therapy.


Assuntos
Fibronectinas/sangue , Hemostasia , Trombose/sangue , Animais , Coagulação Sanguínea , Plaquetas/fisiologia , Feminino , Fibrina/química , Fibrinogênio/metabolismo , Homeostase , Humanos , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Varredura , Resultado do Tratamento
6.
Langmuir ; 30(11): 3217-22, 2014 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-24625256

RESUMO

In contemporary society, a large percentage of medical equipment coming in contact with blood is manufactured from plastic polymers. Unfortunately, exposure may result in undesirable protein-material interactions that can potentially trigger deleterious biological processes such as thrombosis. To address this problem, we have developed an ultrathin antithrombogenic coating based on monoethylene glycol silane surface chemistry. The strategy is exemplified with polycarbonate--a plastic polymer increasingly employed in the biomedical industry. The various straightforward steps of surface modification were characterized with X-ray photoelectron spectroscopy supplemented by contact angle goniometry. Antithrombogenicity was assessed after 5 min exposure to whole human blood dispensed at a shear rate of 1000 s(-1). Remarkably, platelet adhesion, aggregation, and thrombus formation on the coated surface was greatly inhibited (>97% decrease in surface coverage) compared to the bare substrate and, most importantly, nearly nonexistent.


Assuntos
Etilenoglicol/química , Plásticos/química , Plásticos/farmacologia , Silanos/química , Trombose/prevenção & controle , Compostos Benzidrílicos/química , Humanos , Fenóis/química , Plásticos/toxicidade , Cimento de Policarboxilato/química , Propriedades de Superfície
8.
Thromb Haemost ; 111(2): 279-89, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24172860

RESUMO

Platelet adhesion and aggregation at the sites of vascular injury are key events for thrombosis and haemostasis. It has been well demonstrated that interaction between glycoprotein (GP) Ibα and von Willebrand factor (VWF) initiates platelet adhesion and contributes to platelet aggregation, particularly at high shear. GPIb has long been suggested as a desirable antithrombotic target, but anti-GPIb therapy has never been successfully developed. Here, we evaluated the antithrombotic potential of Anfibatide, a novel snake venom-derived GPIb antagonist.We found Anfibatide inhibited washed murine platelet aggregation induced by ristocetin and recombinant murine VWF. It also blocked botrocetin-induced binding of murine plasma VWF to recombinant human GPIbα. Interestingly, Anfibatide did not inhibit botrocetin-induced aggregation of platelet-rich plasma, indicating that its binding site may differ from other snake venom-derived GPIb antagonists. Anfibatide strongly inhibited platelet adhesion, aggregation, and thrombus formation in perfusion chambers at high shear conditions and efficiently dissolved preformed thrombi. Anfibatide also inhibited thrombus growth at low shear conditions, though less than at high shear. Using intravital microscopy, we found that Anfibatide markedly inhibited thrombosis in laser-injured cremaster vessels and prevented vessel occlusion in FeCl3-injured mesenteric vessels. Importantly, Anfibatide further inhibited residual thrombosis in VWF-deficient mice, suggesting that Anfibatide has additional antithrombotic effect beyond its inhibitory role in GPIb-VWF interaction. Anfibatide did not significantly cause platelet activation, prolong tail bleeding time, or cause bleeding diathesis in mice. Thus, consistent with the data from an ongoing clinical trial, the data from this study suggests that Anfibatide is a potent and safe antithrombotic agent.


Assuntos
Plaquetas/efeitos dos fármacos , Venenos de Crotalídeos/farmacologia , Fibrinolíticos/farmacologia , Adesividade Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Complexo Glicoproteico GPIb-IX de Plaquetas/antagonistas & inibidores , Trombose/prevenção & controle , Animais , Sítios de Ligação , Plaquetas/metabolismo , Cloretos , Venenos de Crotalídeos/toxicidade , Modelos Animais de Doenças , Feminino , Compostos Férricos , Fibrinolíticos/toxicidade , Hemorragia/induzido quimicamente , Lectinas Tipo C , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/toxicidade , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Ligação Proteica , Trombose/sangue , Trombose/etiologia , Trombose/genética , Fatores de Tempo , Lesões do Sistema Vascular/complicações , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
9.
Nat Med ; 19(5): 586-94, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23584088

RESUMO

Platelets have a key role in atherogenesis and its complications. Both hypercholesterolemia and increased platelet production promote atherothrombosis; however, a potential link between altered cholesterol homeostasis and platelet production has not been explored. Here we show that transplantation of bone marrow deficient in ABCG4, a transporter of unknown function, into Ldlr(-/-) mice resulted in thrombocytosis, accelerated thrombosis and atherosclerosis. Although not detected in atherosclerotic lesions, Abcg4 was highly expressed in bone marrow megakaryocyte progenitors (MkPs). Abcg4(-/-) MkPs had defective cholesterol efflux to high-density lipoprotein (HDL), increased cell surface expression of the thrombopoietin (TPO) receptor (c-MPL) and enhanced proliferation. These consequences of ABCG4 deficiency seemed to reflect disruption of negative feedback regulation of c-MPL signaling by the E3 ligase c-CBL and the cholesterol-sensing LYN kinase. HDL infusion reduced platelet counts in Ldlr(-/-) mice and in a mouse model of myeloproliferative neoplasm in an ABCG4-dependent fashion. HDL infusions may offer a new approach to reducing atherothrombotic events associated with increased platelet production.


Assuntos
Plaquetas/citologia , Colesterol/metabolismo , Megacariócitos/citologia , Células-Tronco/citologia , Trombocitose/metabolismo , Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Proliferação de Células , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de LDL/genética , Receptores de Trombopoetina/metabolismo , Trombose/patologia , Ubiquitina-Proteína Ligases/metabolismo , Quinases da Família src/metabolismo
10.
Cardiovasc Hematol Disord Drug Targets ; 12(2): 126-32, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23030445

RESUMO

Platelets are small anucleate cells generated from megakaryocytes in the bone marrow. After being released into the circulation, platelets play key roles in the surveillance of vascular injury, and can quickly adhere and aggregate at the site of injury, which are critical events for vascular repair and hemostasis. However, the same biological processes of platelet adhesion and aggregation may also cause thrombotic disorders. The formation of a platelet plug at sites of atherosclerotic lesion rupture is the most common mechanism leading to myocardial or cerebral infarction. Platelet-related deep vein thrombosis is also one of the leading causes of mortality worldwide. The contribution of several platelet receptors and their ligands has been highlighted in these processes. In platelet adhesion, particularly at high shear stress, GPIbα-von Willebrand factor (VWF) interaction may initiate this event, which is followed by GPVI signalling and firm platelet adhesion mediated by members of the integrin family, such as ß3 (αIIbß3) and ß1 (α2ß1, α5ß1) integrins. In platelet aggregation, although GPIbα-VWF, P selectin-sulfatides, and other molecules, may be involved, the process is mainly mediated by ß3 (αIIbß3) integrin and its ligands, such as fibrinogen and VWF. It is intriguing that platelet adhesion and aggregation still occur in mice lacking both fibrinogen and VWF, suggesting that other unforeseen molecule(s) may also be important in these processes. Identification and characterization of these molecules will enrich our knowledge in the basic science of hemostasis and thrombosis, and may lead to the development of new therapies against bleeding disorders and thrombotic diseases.


Assuntos
Plaquetas/patologia , Trombose/sangue , Animais , Hemostasia , Humanos
11.
Circulation ; 126(11): 1385-95, 2012 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-22865890

RESUMO

BACKGROUND: Vascular injury and atherothrombosis involve vessel infiltration by inflammatory leukocytes, migration of medial vascular smooth muscle cells to the intimal layer, and ultimately acute thrombosis. A strategy to simultaneously target these pathological processes has yet to be identified. The secreted protein, Slit2, and its transmembrane receptor, Robo-1, repel neuronal migration in the developing central nervous system. More recently, it has been appreciated that Slit2 impairs chemotaxis of leukocytes and vascular smooth muscle cells toward diverse inflammatory attractants. The effects of Slit2 on platelet function and thrombus formation have never been explored. METHODS AND RESULTS: We detected Robo-1 expression in human and murine platelets and megakaryocytes and confirmed its presence via immunofluorescence microscopy and flow cytometry. In both static and shear microfluidic assays, Slit2 impaired platelet adhesion and spreading on diverse extracellular matrix substrates by suppressing activation of Akt. Slit2 also prevented platelet activation on exposure to ADP. In in vivo studies, Slit2 prolonged bleeding times in murine tail bleeding assays. Using intravital microscopy, we found that after mesenteric arteriolar and carotid artery injury, Slit2 delayed vessel occlusion time and prevented the stable formation of occlusive arteriolar thrombi. CONCLUSIONS: These data demonstrate that Slit2 is a powerful negative regulator of platelet function and thrombus formation. The ability to simultaneously block multiple events in vascular injury may allow Slit2 to effectively prevent and treat thrombotic disorders such as myocardial infarction and stroke.


Assuntos
Plaquetas/fisiologia , Movimento Celular/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Animais , Trombose das Artérias Carótidas/induzido quimicamente , Trombose das Artérias Carótidas/fisiopatologia , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Cloretos/efeitos adversos , Compostos Férricos/efeitos adversos , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , Proteínas do Tecido Nervoso/farmacologia , Adesividade Plaquetária/fisiologia , Receptores Imunológicos/fisiologia , Fatores de Risco , Proteínas Roundabout
12.
PLoS One ; 7(5): e37323, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22624015

RESUMO

Delphinidin-3-glucoside (Dp-3-g) is one of the predominant bioactive compounds of anthocyanins in many plant foods. Although several anthocyanin compounds have been reported to be protective against cardiovascular diseases (CVDs), the direct effect of anthocyanins on platelets, the key players in atherothrombosis, has not been studied. The roles of Dp-3-g in platelet function are completely unknown. The present study investigated the effects of Dp-3-g on platelet activation and several thrombosis models in vitro and in vivo. We found that Dp-3-g significantly inhibited human and murine platelet aggregation in both platelet-rich plasma and purified platelets. It also markedly reduced thrombus growth in human and murine blood in perfusion chambers at both low and high shear rates. Using intravital microscopy, we observed that Dp-3-g decreased platelet deposition, destabilized thrombi, and prolonged the time required for vessel occlusion. Dp-3-g also significantly inhibited thrombus growth in a carotid artery thrombosis model. To elucidate the mechanisms, we examined platelet activation markers via flow cytometry and found that Dp-3-g significantly inhibited the expression of P-selectin, CD63, CD40L, which reflect platelet α- and δ-granule release, and cytosol protein secretion, respectively. We further demonstrated that Dp-3-g downregulated the expression of active integrin αIIbß3 on platelets, and attenuated fibrinogen binding to platelets following agonist treatment, without interfering with the direct interaction between fibrinogen and integrin αIIbß3. We found that Dp-3-g reduced phosphorylation of adenosine monophosphate-activated protein kinase, which may contribute to the observed inhibitory effects on platelet activation. Thus, Dp-3-g significantly inhibits platelet activation and attenuates thrombus growth at both arterial and venous shear stresses, which likely contributes to its protective roles against thrombosis and CVDs.


Assuntos
Antocianinas/farmacologia , Doenças Cardiovasculares/prevenção & controle , Regulação da Expressão Gênica/efeitos dos fármacos , Glucosídeos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Trombose/prevenção & controle , Proteínas Quinases Ativadas por AMP/metabolismo , Análise de Variância , Animais , Tempo de Sangramento , Ligante de CD40/metabolismo , Citometria de Fluxo , Humanos , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Selectina-P/metabolismo , Fosforilação/efeitos dos fármacos , Tetraspanina 30/metabolismo
13.
Arterioscler Thromb Vasc Biol ; 32(7): 1724-31, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22539596

RESUMO

OBJECTIVE: Thrombosis occurs at sites of vascular injury when platelets adhere to subendothelial matrix proteins and to each other. Platelets express many surface receptor proteins, the function of several of these remains poorly characterized. Cadherin 6 is expressed on the platelet surface and contains an arginine-glycine-aspartic acid motif, suggesting that it might have a supportive role in thrombus formation. The aim of this study was to characterize the role of cadherin 6 in platelet function. METHODS AND RESULTS: Platelet aggregation was inhibited by both antibodies and exogenous soluble cadherin 6. Platelet adhesion to immobilized cadherin 6 was inhibited by arginine-glycine-aspartic acid-serine tetrapeptides. Antibodies to α(IIb)ß(3) inhibited platelet adhesion to cadherin 6. Because platelet aggregation occurs in fibrinogen and von Willebrand factor double-deficient mice, we investigated whether cadherin 6 is an alternative ligand for the integrin α(IIb)ß(3). Platelet aggregation in fibrinogen and von Willebrand factor double-deficient mice was significantly inhibited by an antibody to cadherin 6. In flow-based assays, inhibition of cadherin 6 caused a marked reduction in thrombus formation in both human and mouse blood. CONCLUSIONS: This study demonstrates the role of cadherin 6 as a novel ligand for α(IIb)ß(3) and highlights its function in thrombus formation.


Assuntos
Plaquetas/fisiologia , Caderinas/fisiologia , Agregação Plaquetária , Trombose/etiologia , Sequência de Aminoácidos , Animais , Plaquetas/citologia , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Oligopeptídeos/fisiologia , Adesividade Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo
14.
J Clin Invest ; 121(11): 4537-47, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22019589

RESUMO

Fetal and neonatal immune thrombocytopenia (FNIT) is a severe bleeding disorder caused by maternal antibody-mediated destruction of fetal/neonatal platelets. It is the most common cause of severe thrombocytopenia in neonates, but the frequency of FNIT-related miscarriage is unknown, and the mechanism(s) underlying fetal mortality have not been explored. Furthermore, although platelet αIIbß3 integrin and GPIbα are the major antibody targets in immune thrombocytopenia, the reported incidence of anti-GPIbα-mediated FNIT is rare. Here, we developed mouse models of FNIT mediated by antibodies specific for GPIbα and ß3 integrin and compared their pathogenesis. We found, unexpectedly, that miscarriage occurred in the majority of pregnancies in our model of anti-GPIbα-mediated FNIT, which was far more frequent than in anti-ß3-mediated FNIT. Dams with anti-GPIbα antibodies exhibited extensive fibrin deposition and apoptosis/necrosis in their placentas, which severely impaired placental function. Furthermore, anti-GPIbα (but not anti-ß3) antiserum activated platelets and enhanced fibrin formation in vitro and thrombus formation in vivo. Importantly, treatment with either intravenous IgG or a monoclonal antibody specific for the neonatal Fc receptor efficiently prevented anti-GPIbα-mediated FNIT. Thus, the maternal immune response to fetal GPIbα causes what we believe to be a previously unidentified, nonclassical FNIT (i.e., spontaneous miscarriage but not neonatal bleeding) in mice. These results suggest that a similar pathology may have masked the severity and frequency of human anti-GPIbα-mediated FNIT, but also point to possible therapeutic interventions.


Assuntos
Aborto Espontâneo/etiologia , Aborto Espontâneo/prevenção & controle , Histocompatibilidade Materno-Fetal/imunologia , Imunoglobulinas Intravenosas/uso terapêutico , Complexo Glicoproteico GPIb-IX de Plaquetas/imunologia , Receptores Fc/antagonistas & inibidores , Aborto Espontâneo/imunologia , Animais , Plaquetas/imunologia , Modelos Animais de Doenças , Feminino , Antígenos de Histocompatibilidade Classe I/imunologia , Humanos , Integrina beta3/genética , Integrina beta3/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Complexo Glicoproteico GPIb-IX de Plaquetas/genética , Gravidez , Receptores Fc/imunologia , Trombocitopenia Neonatal Aloimune/etiologia , Trombocitopenia Neonatal Aloimune/imunologia
15.
J Clin Invest ; 121(2): 593-603, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21266777

RESUMO

Thrombotic thrombocytopenic purpura (TTP) is a life-threatening disease characterized by systemic microvascular thrombosis caused by adhesion of platelets to ultra-large vWF (ULVWF) multimers. These multimers accumulate because of a deficiency of the processing enzyme ADAMTS13. vWF protein forms long multimers from homodimers that first form through C-terminal disulfide bonds and then join through their N termini by further disulfide bonding. N-acetylcysteine (NAC) is an FDA-approved drug that has long been used to treat chronic obstructive lung disease and acetaminophen toxicity and is known to function in the former disorder by reducing mucin multimers. Here, we examined whether NAC could reduce vWF multimers, which polymerize in a manner similar to mucins. In vitro, NAC reduced soluble plasma-type vWF multimers in a concentration-dependent manner and rapidly degraded ULVWF multimer strings extruded from activated ECs. The effect was preceded by reduction of the intrachain disulfide bond encompassing the platelet-binding A1 domain. NAC also inhibited vWF-dependent platelet aggregation and collagen binding. Injection of NAC into ADAMTS13-deficient mice led to the rapid resolution of thrombi produced by ionophore treatment of the mesenteric venules and reduced plasma vWF multimers. These results suggest that NAC may be a rapid and effective treatment for patients with TTP.


Assuntos
Acetilcisteína/metabolismo , Plasma/metabolismo , Multimerização Proteica , Púrpura Trombocitopênica Trombótica/sangue , Fator de von Willebrand/química , Fator de von Willebrand/metabolismo , Proteína ADAMTS13 , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Antibacterianos/farmacologia , Células Endoteliais/metabolismo , Humanos , Masculino , Metaloendopeptidases/genética , Metaloendopeptidases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agregação Plaquetária/efeitos dos fármacos , Púrpura Trombocitopênica Trombótica/tratamento farmacológico , Ristocetina/farmacologia , Trombose/metabolismo , Trombose/patologia , Fator de von Willebrand/genética
16.
Blood ; 116(18): 3660-8, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20647570

RESUMO

Fetal and neonatal immune thrombocytopenia (FNIT) is a severe bleeding disorder in which maternal antibodies cross the placenta and destroy fetal/neonatal platelets. It has been demonstrated that the neonatal Fc receptor (FcRn) regulates immunoglobulin G (IgG) homeostasis and plays an important role in transplacental IgG transport. However, the role of FcRn in the pathogenesis and therapy of FNIT has not been studied. Here, we developed an animal model of FNIT using combined ß3 integrin-deficient and FcRn-deficient (ß3(-/-)FcRn(-/-)) mice. We found that ß3(-/-)FcRn(-/-) mice are immunoresponsive to ß3(+/+)FcRn(-/-) platelets. The generated antibodies were ß3 integrin specific and were maintained at levels that efficiently induced thrombocytopenia in adult ß3(+/+)FcRn(-/-) mice. FNIT was observed when immunized ß3(-/-)FcRn(+/+) females were bred with ß3(+/+)FcRn(+/+) males, while no FNIT occurred in ß3(-/-)FcRn(-/-) females bred with ß3(+/+)FcRn(-/-) males, suggesting that FcRn is indispensable for the induction of FNIT. We further demonstrated that fetal FcRn was responsible for the transplacental transport of various IgG isotypes. We found that anti-FcRn antibody and intravenous IgG prevented FNIT, and that intravenous IgG ameliorated FNIT through both FcRn-dependent and -independent pathways. Our data suggest that targeting FcRn may be a potential therapy for human FNIT as well as other maternal pathogenic antibody-mediated diseases.


Assuntos
Feto/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Integrina beta3/imunologia , Receptores Fc/imunologia , Trombocitopenia Neonatal Aloimune/imunologia , Animais , Anticorpos/genética , Anticorpos/imunologia , Anticorpos/metabolismo , Plaquetas/imunologia , Regulação para Baixo , Feminino , Feto/fisiopatologia , Deleção de Genes , Antígenos de Histocompatibilidade Classe I/genética , Humanos , Imunoglobulina G/genética , Imunoglobulina G/imunologia , Imunoglobulina G/metabolismo , Imunoglobulinas Intravenosas/uso terapêutico , Integrina beta3/genética , Masculino , Troca Materno-Fetal , Camundongos , Modelos Animais , Placenta/metabolismo , Gravidez , Transporte Proteico , Receptores Fc/genética , Trombocitopenia Neonatal Aloimune/tratamento farmacológico
17.
Thromb Res ; 125(5): e177-83, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20138333

RESUMO

BACKGROUND: Multimerin 1 is a stored platelet and endothelial cell adhesive protein that shows significant conservation. In vitro, multimerin 1 supports platelet adhesion and it also binds to collagen and enhances von Willebrand factor-dependent platelet adhesion to collagen. As selective, multimerin 1 deficient mice have not been generated, we investigated multimerin 1 effects on platelet adhesion using a subpopulation of C57BL/6J mice with tandem deletion of the genes for multimerin 1 and alpha-synuclein, a protein that inhibits alpha-granule release in vitro. We postulated that multimerin 1/alpha-synuclein deficient mice might show impaired platelet adhesive function from multimerin 1 deficiency and increased alpha-granule release from alpha-synuclein deficiency. METHODS: Platelet function was assessed by intravital microscopy, after ferric chloride injury, using untreated and human multimerin 1-transfused multimerin 1/alpha-synuclein deficient mice, and by in vitro assays of adhesion, aggregation and thrombin-induced P-selectin release. RESULTS: Multimerin 1/alpha-synuclein deficient mice showed impaired platelet adhesion and their defective thrombus formation at sites of vessel injury improved with multimerin 1 transfusion. Although multimerin 1/alpha-synuclein deficient platelets showed increased P-selectin release at low thrombin concentrations, they also showed impaired adhesion to collagen, and attenuated aggregation with thrombin, that improved with added multimerin 1. CONCLUSIONS: Our data suggest that multimerin 1 supports platelet adhesive functions and thrombus formation, which will be important to verify by generating and testing selective multimerin 1 deficient mice.


Assuntos
Coagulação Sanguínea/fisiologia , Proteínas Sanguíneas/administração & dosagem , Proteínas Sanguíneas/metabolismo , Adesividade Plaquetária/fisiologia , alfa-Sinucleína/metabolismo , Animais , Coagulação Sanguínea/efeitos dos fármacos , Proteínas Sanguíneas/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Adesividade Plaquetária/efeitos dos fármacos , alfa-Sinucleína/genética
18.
Blood ; 113(8): 1809-17, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19036705

RESUMO

We previously showed that platelet aggregation and thrombus formation occurred in mice lacking both fibrinogen (Fg) and von Willebrand factor (VWF) and that plasma fibronectin (pFn) promoted thrombus growth and stability in injured arterioles in wild-type mice. To examine whether pFn is required for Fg/VWF-independent thrombosis, we generated Fg/VWF/conditional pFn triple-deficient (TKO; Cre(+), Fn(flox/flox), Fg/VWF(-/-)) mice and littermate control (Cre(-), Fn(flox/flox), Fg/VWF(-/-)) mice. Surprisingly, TKO platelet aggregation was not abolished, but instead was enhanced in both heparinized platelet-rich plasma and gel-filtered platelets. This enhancement was diminished when TKO platelets were aggregated in pFn-positive control platelet-poor plasma (PPP), whereas aggregation was enhanced when control platelets were aggregated in pFn-depleted TKO PPP. The TKO platelet aggregation can be completely inhibited by our newly developed mouse anti-mouse beta(3) integrin antibodies but was not affected by anti-mouse GPIbalpha antibodies. Enhanced platelet aggregation was also observed when heparinized TKO blood was perfused in collagen-coated perfusion chambers. Using intravital microscopy, we further showed that thrombogenesis in TKO mice was enhanced in both FeCl(3)-injured mesenteric arterioles and laser-injured cremaster arterioles. Our data indicate that pFn is not essential for Fg/VWF-independent thrombosis and that soluble pFn is probably an important inhibitory factor for platelet aggregation.


Assuntos
Fibrinogênio/genética , Fibronectinas/sangue , Fibronectinas/genética , Agregação Plaquetária/fisiologia , Trombose/metabolismo , Fator de von Willebrand/genética , Animais , Fibrinogênio/metabolismo , Técnicas In Vitro , Integrases/genética , Artérias Mesentéricas/lesões , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Músculo Esquelético/irrigação sanguínea , Adesividade Plaquetária/fisiologia , Contagem de Plaquetas , Solubilidade , Trombose/fisiopatologia , Fator de von Willebrand/metabolismo
19.
Blood ; 113(8): 1818-28, 2009 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-19008452

RESUMO

Carcinoembryonic antigen cell adhesion molecule-1 (CEACAM1) is a surface glycoprotein expressed on various blood cells, epithelial cells, and vascular cells. CEACAM1 possesses adhesive and signaling properties mediated by its intrinsic immunoreceptor tyrosine-based inhibitory motifs that recruit SHP-1 protein-tyrosine phosphatase. In this study, we demonstrate that CEACAM1 is expressed on the surface and in intracellular pools of platelets. In addition, CEACAM1 serves to negatively regulate signaling of platelets by collagen through the glycoprotein VI (GPVI)/Fc receptor (FcR)-gamma-chain. ceacam1(-/-) platelets displayed enhanced type I collagen and GPVI-selective ligand, collagen-related peptide (CRP), CRP-mediated platelet aggregation, enhanced platelet adhesion on type I collagen, and elevated CRP-mediated alpha and dense granule secretion. Platelets derived from ceacam1(-/-) mice form larger thrombi when perfused over a collagen matrix under arterial flow compared with wild-type mice. Furthermore, using intravital microscopy to ferric chloride-injured mesenteric arterioles, we show that thrombi formed in vivo in ceacam1(-/-) mice were larger and were more stable than those in wild-type mice. GPVI depletion using monoclonal antibody JAQ1 treatment of ceacam1(-/-) mice showed a reversal in the more stable thrombus growth phenotype. ceacam1(-/-) mice were more susceptible to type I collagen-induced pulmonary thromboembolism than wild-type mice. Thus, CEACAM1 acts as a negative regulator of platelet-collagen interactions and of thrombus growth involving the collagen GPVI receptor in vitro and in vivo.


Assuntos
Plaquetas/metabolismo , Antígeno Carcinoembrionário/genética , Antígeno Carcinoembrionário/metabolismo , Colágeno Tipo I/metabolismo , Trombose/metabolismo , Animais , Retroalimentação Fisiológica/fisiologia , Feminino , Hematopoese/fisiologia , Homeostase/fisiologia , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fosfolipase C beta/metabolismo , Fosforilação/fisiologia , Adesividade Plaquetária/fisiologia , Glicoproteínas da Membrana de Plaquetas/agonistas , Glicoproteínas da Membrana de Plaquetas/metabolismo , Embolia Pulmonar/metabolismo , Embolia Pulmonar/fisiopatologia , Trombose/fisiopatologia
20.
Thromb Res ; 121(2): 225-34, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17490728

RESUMO

INTRODUCTION: Recently, in vitro models of coagulation have called into question the traditional conception of Factor IX as an intrinsic pathway protein, essential to propagation of coagulation but not central to the initiation of hemostatic plug, which has been thought instead to involve TF/FVIIa interactions with factor X and platelets. We hypothesized that the activation of factor IX, and its role in a factor IXa/FVIIa "tenase" complex leading to thrombin generation, plays a more important role than that of TF/FVIIa complex activation of factor X in the early hemostatic response to vascular injury. In vivo modeling is possible because of the generation of factor IX(-/-) mice. MATERIALS AND METHODS: We used two models of arterial vascular injury, histological examination following mechanical carotid artery disruption and intravital microscopy of a mesenteric arteriole subsequent to ferric chloride arteriolar injury to examine mice having complete deficiency of factor IX (FIX(-/-)). RESULTS: Both injury models demonstrate that platelet rich thrombi /hemostatic plug in FIX(-/-) mice is dramatically reduced as compared to wild type mice under conditions of high shear; in fact, no platelet thrombi (>20 mum) were observed in the intravital experiments. Interestingly, the platelet defect is more striking than that described in mice lacking fibrinogen and/or von Willebrand factor. CONCLUSIONS: The results suggest TF/FVIIa-->FX pathway is insufficient for effective platelet aggregation in the presence of high flow, requiring factor IX at the convergence of both intrinsic and extrinsic pathways. Following platelet adhesion, factor IX is required for normal platelet aggregation in vivo, as well as thrombin generation and propagation of occlusive thrombus at the site of vascular injury.


Assuntos
Vasos Sanguíneos/fisiopatologia , Lesões das Artérias Carótidas/fisiopatologia , Fator IX/genética , Hemofilia B/genética , Trombose/genética , Animais , Vasos Sanguíneos/lesões , Modelos Animais de Doenças , Endotélio Vascular/lesões , Endotélio Vascular/fisiopatologia , Camundongos , Camundongos Knockout , Agregação Plaquetária/genética , Agregação Plaquetária/fisiologia , Trombose/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...