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










Base de datos
Intervalo de año de publicación
1.
PLoS One ; 13(1): e0190728, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29293656

RESUMEN

AIMS: Medical treatment of arterial thrombosis is mainly directed against platelets and coagulation factors, and can lead to bleeding complications. Novel antithrombotic therapies targeting immune cells and neutrophil extracellular traps (NETs) are currently being investigated in animals. We addressed whether immune cell composition of arterial thrombi induced in mouse models of thrombosis resemble those of human patients with acute myocardial infarction (AMI). METHODS AND RESULTS: In a prospective cohort study of patients suffering from AMI, 81 human arterial thrombi were harvested during percutaneous coronary intervention and subjected to detailed histological analysis. In mice, arterial thrombi were induced using two distinct experimental models, ferric chloride (FeCl3) and wire injury of the carotid artery. We found that murine arterial thrombi induced by FeCl3 were highly concordant with human coronary thrombi regarding their immune cell composition, with neutrophils being the most abundant cell type, as well as the presence of NETs and coagulation factors. Pharmacological treatment of mice with the protein arginine deiminase (PAD)-inhibitor Cl-amidine abrogated NET formation, reduced arterial thrombosis and limited injury in a model of myocardial infarction. CONCLUSIONS: Neutrophils are a hallmark of arterial thrombi in patients suffering from acute myocardial infarction and in mouse models of arterial thrombosis. Inhibition of PAD could represent an interesting strategy for the treatment of arterial thrombosis to reduce neutrophil-associated tissue damage and improve functional outcome.


Asunto(s)
Modelos Animales de Enfermedad , Infarto del Miocardio/patología , Ornitina/análogos & derivados , Trombosis/patología , Anciano , Animales , Cloruros/administración & dosificación , Vasos Coronarios/patología , Femenino , Compuestos Férricos/administración & dosificación , Humanos , Masculino , Ratones , Persona de Mediana Edad , Ornitina/farmacología , Estudios Prospectivos
2.
PLoS One ; 12(3): e0172788, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28253287

RESUMEN

Platelets modulate the process of cancer metastasis. However, current knowledge on the direct interaction of platelets and tumor cells is mostly based on findings obtained in vitro. We addressed the role of the platelet fibrinogen receptor glycoprotein IIb (integrin αIIb) for experimental melanoma metastasis in vivo. Highly metastatic B16-D5 melanoma cells were injected intravenously into GPIIb-deficient (GPIIb-/-) or wildtype (WT) mice. Acute accumulation of tumor cells in the pulmonary vasculature was assessed in real-time by confocal videofluorescence microscopy. Arrest of tumor cells was dramatically reduced in GPIIb-/- mice as compared to WT. Importantly, we found that mainly multicellular aggregates accumulated in the pulmonary circulation of WT, instead B16-D5 aggregates were significantly smaller in GPIIb-/- mice. While pulmonary arrest of melanoma was clearly dependent on GPIIb in this early phase of metastasis, we also addressed tumor progression 10 days after injection. Inversely, and unexpectedly, we found that melanoma metastasis was now increased in GPIIb-/- mice. In contrast, GPIIb did not regulate local melanoma proliferation in a subcutaneous tumor model. Our data suggest that the platelet fibrinogen receptor has a differential role in the modulation of hematogenic melanoma metastasis. While platelets clearly support early steps in pulmonary metastasis via GPIIb-dependent formation of platelet-tumor-aggregates, at a later stage its absence is associated with an accelerated development of melanoma metastases.


Asunto(s)
Plaquetas/metabolismo , Neoplasias Pulmonares/secundario , Pulmón/patología , Melanoma Experimental/patología , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Animales , Plaquetas/fisiología , Agregación Celular , Línea Celular Tumoral , Proliferación Celular , Pulmón/irrigación sanguínea , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/irrigación sanguínea , Neoplasias Pulmonares/patología , Ratones , Microcirculación
3.
Sci Transl Med ; 8(367): 367ra168, 2016 11 30.
Artículo en Inglés | MEDLINE | ID: mdl-27903864

RESUMEN

In patients with atrial fibrillation, oral anticoagulation with oral thrombin inhibitors (OTIs), in contrast to vitamin K antagonists (VKAs), associates with a modest increase in acute coronary syndromes (ACSs). Whether this observation is causatively linked to OTI treatment and, if so, whether OTI action is the result of a lower antithrombotic efficacy of OTI compared to VKA or reflects a yet undefined prothrombotic activity of OTI remain unclear. We analyzed platelet function in patients receiving OTI or dose-adapted VKA under static and flow conditions. In vivo, we studied arterial thrombosis in OTI-, VKA-, and vehicle-treated mice using carotid ligation and wire injury models. Further, we examined thrombus formation on human atherosclerotic plaque homogenates under arterial shear to address the relevance to human pathology. Under static conditions, aggregation in the presence of ristocetin was increased in OTI-treated blood, whereas platelet reactivity and aggregation to other agonists were only marginally affected. Under flow conditions, firm platelet adhesion and thrombus formation on von Willebrand factor, collagen, and human atherosclerotic plaque were increased in the presence of OTI in comparison to VKA. OTI treatment was associated with increased thrombus formation in injured carotid arteries of mice. Inhibition or ablation of GPIbα-thrombin interactions abolished the effect of OTI on thrombus formation, suggesting a mechanistic role of the platelet receptor GPIbα and its thrombin-binding site. The effect of OTI was also abrogated in the presence of aspirin. In summary, OTI treatment has prothrombotic activity that might contribute to the increase in ACS observed clinically in patients.


Asunto(s)
Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Trombina/antagonistas & inhibidores , Trombosis/patología , Síndrome Coronario Agudo/patología , Administración Oral , Animales , Anticoagulantes/farmacología , Arterias/patología , Aspirina/farmacología , Aterosclerosis/patología , Sitios de Unión , Plaquetas/efectos de los fármacos , Fibrinolíticos/farmacología , Humanos , Ratones , Inhibidores de Agregación Plaquetaria/farmacología , Ensayos Clínicos Controlados Aleatorios como Asunto , Vitamina K/antagonistas & inhibidores
4.
Arterioscler Thromb Vasc Biol ; 35(3): 589-97, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25550202

RESUMEN

OBJECTIVE: Arteriogenesis is strongly dependent on the recruitment of leukocytes, especially monocytes, into the perivascular space of growing collateral vessels. On the basis of previous findings that platelets are central players in inflammatory processes and mediate the recruitment of leukocytes, the aim of this study was to assess the role of platelets in a model of arterial remodeling. APPROACH AND RESULTS: C57Bl6 wild-type mice, IL4-R/Iba mice lacking the extracellular domain of the glycoprotein Ibα (GPIbα) receptor, and mice treated with antibodies to block GPIbα or deplete circulating platelets were studied in peripheral arteriogenesis. Using a novel model of intravital 2-photon and epifluorescence imaging, we visualized and quantified the interaction of platelets with leukocytes and the vascular endothelium in vivo. We found that transient platelet adhesion to the endothelium of collateral vessels was a major event during arteriogenesis and depended on GPIbα. Furthermore, leukocyte recruitment was obviously affected in animals with defective platelet GPIbα function. In IL4-R/Iba mice, transient and firm leukocyte adhesion to the endothelium of collateral vessels, as well as leukocyte accumulation in the perivascular space, were significantly reduced. Furthermore, we detected platelet-leukocyte aggregates within the circulation, which were significantly reduced in IL4-R/Iba animals. Finally, platelet depletion and loss of GPIbα function resulted in poor reperfusion recovery as determined by laser Doppler imaging. CONCLUSIONS: Thus, GPIbα-mediated interactions between platelets and endothelial cells, as well as leukocytes, support innate immune cell recruitment and promote arteriogenesis-establishing platelets as critical players in this process.


Asunto(s)
Neovascularización Fisiológica , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Animales
5.
Am J Physiol Cell Physiol ; 303(7): C757-66, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22814400

RESUMEN

Platelets play a critical role in the pathophysiology of reperfusion, sepsis, and cardiovascular diseases. In a multiple step process, they adhere to activated endothelium and release proinflammatory cytokines thereby promoting the inflammatory process. Glycoprotein VI (GPVI) is the major collagen receptor on the platelet surface and triggers platelet activation and primary hemostasis. Activation of GPVI leads to stable platelet adhesion and degranulation of platelet granules. However, GPVI is critically involved in platelet adhesion to activated endothelium without exposure of subendothelial matrix. Earlier studies show that the soluble GPVI-Fc binds to collagen and protects mice from atherosclerosis and decreases neointima proliferation after arterial injury. Here, we show for the first time that recombinant GPVI-Fc binds to activated endothelium mainly via vitronectin and prevents platelet/endothelial interaction. Administration of GPVI-Fc reduced infarct size and preserved cardiac function in a mouse model of myocardial infarction. This process was associated with reduced GPVI-induced platelet degranulation and release of proinflammatory cytokines in vitro and in vivo. Taken together, administration of GPVI-Fc offers a novel strategy to control platelet-mediated inflammation and to preserve myocardial function following myocardial infarction.


Asunto(s)
Endotelio Vascular/metabolismo , Corazón/fisiología , Fragmentos Fc de Inmunoglobulinas/metabolismo , Ataque Isquémico Transitorio/metabolismo , Adhesividad Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Animales , Células CHO , Bovinos , Cricetinae , Cricetulus , Endotelio Vascular/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Multimerización de Proteína/fisiología
6.
J Exp Med ; 209(4): 819-35, 2012 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-22451716

RESUMEN

Deep vein thrombosis (DVT) is a major cause of cardiovascular death. The sequence of events that promote DVT remains obscure, largely as a result of the lack of an appropriate rodent model. We describe a novel mouse model of DVT which reproduces a frequent trigger and resembles the time course, histological features, and clinical presentation of DVT in humans. We demonstrate by intravital two-photon and epifluorescence microscopy that blood monocytes and neutrophils crawling along and adhering to the venous endothelium provide the initiating stimulus for DVT development. Using conditional mutants and bone marrow chimeras, we show that intravascular activation of the extrinsic pathway of coagulation via tissue factor (TF) derived from myeloid leukocytes causes the extensive intraluminal fibrin formation characteristic of DVT. We demonstrate that thrombus-resident neutrophils are indispensable for subsequent DVT propagation by binding factor XII (FXII) and by supporting its activation through the release of neutrophil extracellular traps (NETs). Correspondingly, neutropenia, genetic ablation of FXII, or disintegration of NETs each confers protection against DVT amplification. Platelets associate with innate immune cells via glycoprotein Ibα and contribute to DVT progression by promoting leukocyte recruitment and stimulating neutrophil-dependent coagulation. Hence, we identified a cross talk between monocytes, neutrophils, and platelets responsible for the initiation and amplification of DVT and for inducing its unique clinical features.


Asunto(s)
Plaquetas/fisiología , Comunicación Celular , Monocitos/fisiología , Neutrófilos/fisiología , Trombosis de la Vena/etiología , Animales , Factor XII/metabolismo , Ratones , Ratones Endogámicos C57BL , Selectina-P/fisiología , Tromboplastina/fisiología
7.
Nat Med ; 16(8): 887-96, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20676107

RESUMEN

Blood neutrophils provide the first line of defense against pathogens but have also been implicated in thrombotic processes. This dual function of neutrophils could reflect an evolutionarily conserved association between blood coagulation and antimicrobial defense, although the molecular determinants and in vivo significance of this association remain unclear. Here we show that major microbicidal effectors of neutrophils, the serine proteases neutrophil elastase and cathepsin G, together with externalized nucleosomes, promote coagulation and intravascular thrombus growth in vivo. The serine proteases and extracellular nucleosomes enhance tissue factor- and factor XII-dependent coagulation in a process involving local proteolysis of the coagulation suppressor tissue factor pathway inhibitor. During systemic infection, activation of coagulation fosters compartmentalization of bacteria in liver microvessels and reduces bacterial invasion into tissue. In the absence of a pathogen challenge, neutrophil-derived serine proteases and nucleosomes can contribute to large-vessel thrombosis, the main trigger of myocardial infarction and stroke. The ability of coagulation to suppress pathogen dissemination indicates that microvessel thrombosis represents a physiological tool of host defense.


Asunto(s)
Coagulación Sanguínea/genética , Inmunidad Innata/genética , Neutrófilos/fisiología , Serina Proteasas/fisiología , Animales , Coagulación Sanguínea/fisiología , Factores de Coagulación Sanguínea/metabolismo , Factores de Coagulación Sanguínea/fisiología , Catepsina G/genética , Catepsina G/metabolismo , Catepsina G/fisiología , Fibrina/metabolismo , Inmunidad Innata/fisiología , Elastasa de Leucocito/genética , Elastasa de Leucocito/metabolismo , Elastasa de Leucocito/fisiología , Lipoproteínas/metabolismo , Ratones , Ratones Noqueados , Modelos Biológicos , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Neutrófilos/metabolismo , Nucleosomas/metabolismo , Procesamiento Proteico-Postraduccional/genética , Procesamiento Proteico-Postraduccional/fisiología , Serina Proteasas/genética , Serina Proteasas/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/metabolismo
8.
J Mol Cell Cardiol ; 49(3): 532-42, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20430036

RESUMEN

Glycoprotein VI (GPVI) mediates binding of platelets to subendothelial collagen during acute arterial thrombosis. GPVI interactions with the activated atherosclerotic vascular endothelium during early atherosclerosis, however, are not well understood. In ApoE-/- mice, platelet adhesion to atherosclerotic arteries was increased, as measured by intravital microscopy. This platelet adhesion was significantly inhibited by IV injection of GPVI-Fc (1 mg/kg body weight). Atherosclerosis in ApoE-/- mice was attenuated both after 7 and 10 weeks of treatment with the anti-GPVI antibody JAQ1 (2 mg/kg body weight i.p. twice weekly). Binding of GPVI-Fc (1 mg/kg IV) occurred to deeper layers, but also to the luminal site of plaques in atherosclerotic rabbits, but not to the vessel wall of healthy littermates. Gene transfer of GPVI-Fc to the carotid vascular wall significantly attenuated athero-progression and endothelial dysfunction in atherosclerotic rabbits in vivo. Specific binding of the soluble GPVI receptor (GPVI-Fc) to fibronectin was found in vitro to coated ELISA plates. Platelet adhesion to fibronectin was significantly inhibited both by GPVI-Fc and by the anti-GPVI antibody 5C4 ex vivo in flow chamber experiments. GPVI plays a role in platelet adhesion to atherosclerotic endothelium in the absence of plaque rupture. Inhibition of GPVI both via GPVI-Fc and anti-GPVI-antibodies results in protection against atherosclerosis in both cholesterol-fed rabbits and ApoE-/- mice. This novel mechanism of GPVI-mediated platelet adhesion-possibly via fibronectin-could relevantly contribute to platelet-triggered atheroprogression.


Asunto(s)
Aterosclerosis/patología , Endotelio Vascular/metabolismo , Fibronectinas/metabolismo , Adhesividad Plaquetaria/fisiología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Adenoviridae/genética , Animales , Anticuerpos Monoclonales/farmacología , Apolipoproteínas E/fisiología , Aterosclerosis/metabolismo , Células CHO , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Cricetinae , Cricetulus , Endotelio Vascular/patología , Técnicas de Transferencia de Gen , Proteínas Fluorescentes Verdes , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/inmunología , Conejos
9.
J Clin Invest ; 118(3): 1110-22, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18274674

RESUMEN

The activation of initiator protein tissue factor (TF) is likely to be a crucial step in the blood coagulation process, which leads to fibrin formation. The stimuli responsible for inducing TF activation are largely undefined. Here we show that the oxidoreductase protein disulfide isomerase (PDI) directly promotes TF-dependent fibrin production during thrombus formation in vivo. After endothelial denudation of mouse carotid arteries, PDI was released at the injury site from adherent platelets and disrupted vessel wall cells. Inhibition of PDI decreased TF-triggered fibrin formation in different in vivo murine models of thrombus formation, as determined by intravital fluorescence microscopy. PDI infusion increased - and, under conditions of decreased platelet adhesion, PDI inhibition reduced - fibrin generation at the injury site, indicating that PDI can directly initiate blood coagulation. In vitro, human platelet-secreted PDI contributed to the activation of cryptic TF on microvesicles (microparticles). Mass spectrometry analyses indicated that part of the extracellular cysteine 209 of TF was constitutively glutathionylated. Mixed disulfide formation contributed to maintaining TF in a state of low functionality. We propose that reduced PDI activates TF by isomerization of a mixed disulfide and a free thiol to an intramolecular disulfide. Our findings suggest that disulfide isomerases can act as injury response signals that trigger the activation of fibrin formation following vessel injury.


Asunto(s)
Coagulación Sanguínea , Fibrina/biosíntesis , Proteína Disulfuro Isomerasas/fisiología , Transducción de Señal/fisiología , Tromboplastina/fisiología , Animales , Células Cultivadas , Disulfuros/química , Glutatión/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Tromboplastina/química
10.
Thromb Haemost ; 99(1): 190-5, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18217153

RESUMEN

Acetylsalicylic acid (ASA) and the thienopyridine clopidogrel are established anti-platelet drugs that significantly reduce secondary cardiovascular events in patients with manifest atherosclerosis. However, their impact on atherosclerotic lesion development remains controversial. Four-week-old ApoE-deficient mice were randomly assigned to four groups receiving a cholesterol diet together with either ASA (5 mg/kg), or clopidogrel (25 mg/kg), or a combination of both ASA and clopidogrel, or vehicle for 8-12 weeks. Using intravital microscopy we found that daily administration of ASA in combination with clopidogrel reduces platelet thrombus formation following rupture of atherosclerotic plaque in vivo by approximately 50%. However, therapy with ASA or clopidogrel alone, or in combination for a period of 8-12 weeks had no significant effect on adhesion of platelets to dysfunctional endothelial cells or on atherosclerotic lesion formation in the aortic root or the carotid artery. In conclusion, anti-platelet therapy is effective in reducing platelet adhesion and subsequent thrombus formation following rupture of atherosclerotic plaque in vivo. However, our data do not support a role of either drug in the primary prevention of atherosclerosis in ApoE-deficient mice.


Asunto(s)
Apolipoproteínas E/metabolismo , Aspirina/farmacología , Aterosclerosis/prevención & control , Plaquetas/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/farmacología , Trombosis/prevención & control , Ticlopidina/análogos & derivados , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aspirina/administración & dosificación , Aterosclerosis/sangre , Aterosclerosis/complicaciones , Aterosclerosis/etiología , Aterosclerosis/patología , Plaquetas/metabolismo , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/metabolismo , Colesterol en la Dieta , Clopidogrel , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Esquema de Medicación , Quimioterapia Combinada , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Microscopía por Video , Adhesividad Plaquetaria/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Inhibidores de Agregación Plaquetaria/administración & dosificación , Rotura , Trombosis/sangre , Trombosis/etiología , Trombosis/patología , Ticlopidina/administración & dosificación , Ticlopidina/farmacología , Factores de Tiempo
11.
Arterioscler Thromb Vasc Biol ; 27(5): 1184-90, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17347483

RESUMEN

OBJECTIVE: Thrombus formation after atherosclerotic plaque rupture critically involves the platelet collagen receptor glycoprotein (GP) VI. We investigated the impact of EXP3179, an active metabolite of the angiotensin II type 1 (AT1)-receptor antagonist Losartan (LOS) on GPVI-dependent platelet activation. METHODS AND RESULTS: EXP3179 and LOS but not EXP3174--the major AT1-receptor blocking metabolite of LOS--dose-dependently inhibited collagen-I (P<0.01) and GPVI-dependent platelet aggregation (P<0.01) analyzed by optical aggregometry. Platelet activation was further determined by flow cytometry measuring the expression of platelet PAC-1, an epitope of the activated fibrinogen-receptor complex. EXP3179 and LOS inhibited collagen-I (P<0.01) and GPVI-dependent PAC-1 expression (P<0.01). EXP3179 and LOS but not EXP3174 decreased the adhesion of GPVI-receptor expressing Chinese hamster ovarian cells on collagen-I under arterial shear conditions determined by flow chamber analysis (P<0.01 and P<0.05). EXP3179 also reduced human atherosclerotic plaque material-induced platelet aggregation (P<0.01) in vitro and murine platelet adhesion after acute vessel injury in vivo as determined by intravital microscopy (P<0.01). CONCLUSION: EXP3179 acts as a specific inhibitor of the platelet collagen receptor GPVI independent of AT1-receptor antagonism. Further investigations may clarify its individual potential as a novel pharmacological approach to specifically inhibit atherothrombotic events by GPVI-receptor blockade.


Asunto(s)
Aterosclerosis/complicaciones , Losartán/análogos & derivados , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Trombosis , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Aterosclerosis/sangre , Citometría de Flujo , Humanos , Losartán/uso terapéutico , Microscopía Fluorescente , Glicoproteínas de Membrana Plaquetaria/antagonistas & inhibidores , Receptor de Angiotensina Tipo 1/efectos de los fármacos , Trombosis/sangre , Trombosis/etiología , Trombosis/prevención & control , Resultado del Tratamiento
12.
Blood ; 109(2): 552-9, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16990611

RESUMEN

Defective regulation of platelet activation/aggregation is a predominant cause for arterial thrombosis, the major complication of atherosclerosis triggering myocardial infarction and stroke. A central regulatory pathway conveying inhibition of platelet activation/aggregation is nitric oxide (NO)/cyclic GMP (cGMP) signaling by cGMP-dependent protein kinase I (cGKI). However, the regulatory cascade downstream of cGKI mediating platelet inhibition is still unclear. Here, we show that the inositol-1,4,5-trisphosphate receptor-associated cGMP kinase substrate (IRAG) is abundantly expressed in platelets and assembled in a macrocomplex together with cGKIbeta and the inositol-1,4,5-trisphosphate receptor type I (InsP3RI). cGKI phosphorylates IRAG at Ser664 and Ser677 in intact platelets. Targeted deletion of the IRAG-InsP3RI interaction in IRAGDelta12/Delta12 mutant mice leads to a loss of NO/cGMP-dependent inhibition of fibrinogen-receptor activation and platelet aggregation. Intracellular calcium transients were not affected by DEA/NO or cGMP in mutant platelets. Furthermore, intravital microscopy shows that NO fails to prevent arterial thrombosis of the injured carotid artery in IRAGDelta12/Delta12 mutants. These findings reveal that interaction between IRAG and InsP3RI has a central role in NO/cGMP-dependent inhibition of platelet aggregation and in vivo thrombosis.


Asunto(s)
GMP Cíclico/metabolismo , Proteínas de la Membrana/fisiología , Óxido Nítrico/metabolismo , Fosfoproteínas/fisiología , Agregación Plaquetaria/fisiología , Transducción de Señal/fisiología , Trombosis/prevención & control , Animales , Calcio/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Activadores de Enzimas/farmacología , Humanos , Receptores de Inositol 1,4,5-Trifosfato/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Complejos Multiproteicos/metabolismo , Fosfoproteínas/genética , Fosforilación , Agregación Plaquetaria/efectos de los fármacos , Valores de Referencia , Transducción de Señal/efectos de los fármacos , Trombosis/metabolismo
13.
J Exp Med ; 203(5): 1221-33, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16618794

RESUMEN

The accumulation of smooth muscle and endothelial cells is essential for remodeling and repair of injured blood vessel walls. Bone marrow-derived progenitor cells have been implicated in vascular repair and remodeling; however, the mechanisms underlying their recruitment to the site of injury remain elusive. Here, using real-time in vivo fluorescence microscopy, we show that platelets provide the critical signal that recruits CD34+ bone marrow cells and c-Kit+ Sca-1+ Lin- bone marrow-derived progenitor cells to sites of vascular injury. Correspondingly, specific inhibition of platelet adhesion virtually abrogated the accumulation of both CD34+ and c-Kit+ Sca-1+ Lin- bone marrow-derived progenitor cells at sites of endothelial disruption. Binding of bone marrow cells to platelets involves both P-selectin and GPIIb integrin on platelets. Unexpectedly, we found that activated platelets secrete the chemokine SDF-1alpha, thereby supporting further primary adhesion and migration of progenitor cells. These findings establish the platelet as a major player in the initiation of vascular remodeling, a process of fundamental importance for vascular repair and pathological remodeling after vascular injury.


Asunto(s)
Plaquetas/metabolismo , Células de la Médula Ósea/metabolismo , Movimiento Celular , Quimiocinas CXC/metabolismo , Células Madre/metabolismo , Trombosis/metabolismo , Animales , Antígenos de Diferenciación/metabolismo , Arterias/lesiones , Arterias/metabolismo , Arterias/patología , Plaquetas/patología , Células de la Médula Ósea/patología , Adhesión Celular , Quimiocina CXCL12 , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Ratones , Microscopía Fluorescente , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Activación Plaquetaria , Recuperación de la Función , Células Madre/patología , Trombosis/patología
14.
Circulation ; 112(8): 1180-8, 2005 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-16103235

RESUMEN

BACKGROUND: The platelet glycoprotein (GP) IIb/IIIa integrin binds to fibrinogen and thereby mediates platelet aggregation. Here, we addressed the role of GP IIb for platelet adhesion and determined the relevance of platelet GP IIb for the processes of atherosclerosis and cerebral ischemia-reperfusion (I/R) injury. METHODS AND RESULTS: GP IIb(-/-) mice were generated and bred with ApoE(-/-) animals to create GP IIb(-/-)ApoE(-/-) mice. Platelet adhesion to the mechanically injured or atherosclerotic vessel wall was monitored by in vivo video fluorescence microscopy. In the presence of GP IIb, vascular injury and early atherosclerosis induced platelet adhesion in the carotid artery (CA). In contrast, platelet adhesion was significantly reduced in the absence of GP IIb integrin (P<0.05). To address the contribution of platelet GP IIb to atheroprogression, we determined atherosclerotic lesion formation in the CA and aortic arch (AA) of GP IIb(+/+)ApoE(-/-) or GP IIb(-/-)ApoE(-/-) mice. Interestingly, the absence of GP IIb attenuated lesion formation in CA and AA, indicating that platelets, via GP IIb, contribute substantially to atherosclerosis. Next, we assessed the implication of GP IIb for cerebral I/R injury. We observed that after occlusion of the middle cerebral artery, the cerebral infarct size was drastically reduced in mice lacking GP IIb compared with wild-types. CONCLUSIONS: These findings show for the first time in vivo that GP IIb not only mediates platelet aggregation but also triggers platelet adhesion to exposed extracellular matrices and dysfunctional endothelial cells. In a process strictly involving GP IIb, platelets, which are among the first blood cells to arrive at the scene of endothelial dysfunction, contribute essentially to atherosclerosis and cerebral I/R injury.


Asunto(s)
Isquemia Encefálica/sangre , Arteriosclerosis Intracraneal/sangre , Adhesividad Plaquetaria/fisiología , Glicoproteína IIb de Membrana Plaquetaria/genética , Glicoproteína IIb de Membrana Plaquetaria/metabolismo , Animales , Apolipoproteínas E/genética , Plaquetas/citología , Plaquetas/metabolismo , Isquemia Encefálica/genética , Comunicación Celular/fisiología , Infarto Cerebral/sangre , Infarto Cerebral/genética , Progresión de la Enfermedad , Células Endoteliales/citología , Células Endoteliales/metabolismo , Femenino , Arteriosclerosis Intracraneal/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Agregación Plaquetaria/fisiología , Accidente Cerebrovascular/sangre , Accidente Cerebrovascular/genética
15.
Arterioscler Thromb Vasc Biol ; 25(8): 1750-5, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15933244

RESUMEN

UNLABELLED: Background- We recently reported the development of culture-derived (CD) platelets with the aim to express any protein of interest in these platelets. We now report a specific protocol of retroviral infection into the progenitor cells and subsequent selection, which allows to generate large amounts of highly homogenous transgene-expressing CD platelets and to study transgene function rapidly and reliably at large-scale ex vivo and in vivo settings. METHODS AND RESULTS: After retroviral infection and selection, the activation-dependent expression profile of surface markers, aggregation, and granule release were investigated. The function of transgene-expressing CD platelets, the precursor cells of which had been retrovirally infected, compared well to noninfected CD platelets or freshly isolated platelets. Hence, the retroviral infection protocol did not alter platelet physiology. In contrast, adenoviral infection of precursors to CD platelets resulted in marked functional alterations that obviated their use in analytic experiments. Additionally, sufficient amounts of selected CD platelets were generated to warrant intravenous injections into living mice. This approach permitted study of their adhesive profile at endothelial lesions and their effect on thrombus formation in vivo by intravital videofluorescence microscopy. CONCLUSIONS: The novel selection method allowed us to produce recombinant transgene-expressing platelets in sufficient amounts to study genetically modified platelets in vitro and in vivo.


Asunto(s)
Plaquetas/fisiología , Técnicas de Transferencia de Gen , Retroviridae/genética , Trombosis/fisiopatología , Transgenes/genética , Adenoviridae/genética , Animales , Plaquetas/citología , Células de la Médula Ósea/citología , Células de la Médula Ósea/fisiología , Células Cultivadas , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Humanos , Leucocitos Mononucleares/citología , Leucocitos Mononucleares/fisiología , Megacariocitos/citología , Megacariocitos/fisiología , Ratones , Adhesividad Plaquetaria/fisiología , Proteínas Recombinantes/genética
16.
Thromb Haemost ; 93(5): 910-3, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-15886808

RESUMEN

Glycoprotein VI (GPVI) is the major platelet collagen receptor and plays a critical role in the process of thrombosis at sites of atherosclerotic lesions. This study evaluates the feasibility of radiolabeled soluble GPVI to identify injured arterial lesions. Radiolabeling was carried out using the iodogen method and resulted in the radioiodinated GPVI in radiochemical yields between 97-100%. The biodistribution of [(125)I]GPVI was determined in normal mice and demonstrated a blood clearance halftime of approximately 5.5 hours. Vascular lesions were induced in the carotid artery in wild type and ApoE(-/-)mice. Immediately after injury radioiodinated GPVI was injected intravenously. Binding of [(123)I]GPVI to carotid lesions was assessed by szintigraphic in vivo imaging. Carotid arteries were explanted for ex vivo autoradiography and histological characterization of the lesion. In vivo and ex vivo imaging revealed substantial accumulation of radioiodinated GPVI in the injured artery wall, with a ratio of lesion to control vessel of 3:1 and 7:1, respectively. Because GPVI is the critical collagen receptor that mediates platelet adhesion to vascular lesions, soluble radiolabeled GPVI may be an agent for non-invasive imaging of thrombogenic thus, vulnerable atherosclerotic plaques.


Asunto(s)
Arterias/lesiones , Arterias/patología , Diagnóstico por Imagen/métodos , Glicoproteínas de Membrana Plaquetaria/química , Trombosis/diagnóstico , Animales , Arteriosclerosis/diagnóstico , Arteriosclerosis/patología , Plaquetas/metabolismo , Arterias Carótidas/patología , Dimerización , Masculino , Ratones , Ratones Endogámicos C57BL , Activación Plaquetaria , Adhesividad Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/metabolismo , Glicoproteínas de Membrana Plaquetaria/farmacocinética , Unión Proteica , Receptores de Colágeno/metabolismo , Trombosis/patología , Factores de Tiempo
17.
FASEB J ; 18(2): 397-9, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14656994

RESUMEN

Platelet-collagen interactions play a fundamental role in the process of arterial thrombosis. The major platelet collagen receptor is the glycoprotein VI (GPVI). Here, we determined the effects of a soluble dimeric form of GPVI on platelet adhesion in vitro and in vivo. We fused the extracellular domain of GPVI with the human immunoglobulin Fc domain. The soluble dimeric form of GPVI (GPVI-Fc) specifically bound to immobilized collagen. Binding of GPVI-Fc to collagen was inhibited competitively by soluble GPVI-Fc, but not control Fc lacking the external GPVI domain. GPVI-Fc inhibited the adhesion of CHO cells that stably express human GPVI and of platelets on collagen and attenuated thrombus formation under shear conditions in vitro. To test the effects of GPVI-Fc in vivo, arterial thrombosis was induced in the mouse carotid artery, and platelet-vessel wall interactions were visualized by intravital fluorescence microscopy. Infusion of GPVI-Fc but not of control Fc virtually abolished stable arrest and aggregation of platelets following vascular injury. Importantly, GPVI-Fc but not control Fc, was detected at areas of vascular injury. These findings further substantiate the critical role of the collagen receptor GPVI in the initiation of thrombus formation at sites of vascular injury and identify soluble GPVI as a promising antithrombotic strategy.


Asunto(s)
Plaquetas/fisiología , Arterias Carótidas/metabolismo , Arterias Carótidas/patología , Adhesividad Plaquetaria , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/química , Glicoproteínas de Membrana Plaquetaria/metabolismo , Animales , Tiempo de Sangría , Células CHO , Clonación Molecular , Colágeno/metabolismo , Cricetinae , Dimerización , Humanos , Fragmentos Fc de Inmunoglobulinas/química , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Ratones , Recuento de Plaquetas , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/aislamiento & purificación , Unión Proteica , Reología , Solubilidad , Trombosis
18.
Blood ; 103(1): 136-42, 2004 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12933589

RESUMEN

Platelet adhesion and activation at the vascular wall are the initial steps leading to arterial thrombosis and vascular occlusion. Prostacyclin and nitric oxide inhibit platelet adhesion, acting via cyclic adenosine monophosphate (cAMP)- and cyclic guanosine monophosphate (cGMP)-dependent protein kinases. A major downstream target for both cAMP- and cGMP-dependent protein kinases is the vasodilator-stimulated phosphoprotein (VASP). To test the significance of VASP for the regulation of platelet adhesion in vivo, we studied platelet-vessel wall interactions using VASP-deficient (VASP-/-) mice. Under physiologic conditions, platelet adhesion to endothelial cells was significantly enhanced in VASP null mutants when compared with wild-type mice (P <.05). Platelet recruitment in VASP null mice involved P-selectin and the fibrinogen receptor glycoprotein IIb-IIIa (GPIIb-IIIa). Under pathophysiologic conditions, the loss of VASP increased platelet adhesion to the postischemic intestinal microvasculature, to the atherosclerotic endothelium of ApoE-deficient mice, and to the subendothelial matrix following endothelial denudation (P <.05 vs wild type). Importantly, platelet adhesion in VASP null mutants was unresponsive to nitric oxide. These data show for the first time in vivo that VASP is involved in down-regulation of platelet adhesion to the vascular wall under both physiologic and pathophysiologic conditions.


Asunto(s)
Moléculas de Adhesión Celular/deficiencia , Fosfoproteínas/deficiencia , Adhesividad Plaquetaria/fisiología , Animales , Estenosis Carotídea/patología , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/fisiología , Endotelio Vascular/citología , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos , Donantes de Óxido Nítrico/farmacología , Fosfoproteínas/genética , Fosfoproteínas/fisiología , Adhesividad Plaquetaria/efectos de los fármacos
19.
Nat Med ; 9(11): 1418-22, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14528298

RESUMEN

Platelet activation at sites of vascular injury is essential for primary hemostasis, but also underlies arterial thrombosis leading to myocardial infarction or stroke. Platelet activators such as adenosine diphosphate, thrombin or thromboxane A(2) (TXA(2)) activate receptors that are coupled to heterotrimeric G proteins. Activation of platelets through these receptors involves signaling through G(q), G(i) and G(z) (refs. 4-6). However, the role and relative importance of G(12) and G(13), which are activated by various platelet stimuli, are unclear. Here we show that lack of Galpha(13), but not Galpha(12), severely reduced the potency of thrombin, TXA(2) and collagen to induce platelet shape changes and aggregation in vitro. These defects were accompanied by reduced activation of RhoA and inability to form stable platelet thrombi under high shear stress ex vivo. Galpha(13) deficiency in platelets resulted in a severe defect in primary hemostasis and complete protection against arterial thrombosis in vivo. We conclude that G(13)-mediated signaling processes are required for normal hemostasis and thrombosis and may serve as a new target for antiplatelet drugs.


Asunto(s)
Plaquetas/metabolismo , Subunidades alfa de la Proteína de Unión al GTP G12-G13/metabolismo , Hemostasis/fisiología , Trombosis/metabolismo , Animales , Subunidades alfa de la Proteína de Unión al GTP G12-G13/genética , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Noqueados , Proteínas Virales/genética , Proteínas Virales/metabolismo
20.
J Exp Med ; 197(1): 41-9, 2003 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-12515812

RESUMEN

Platelet adhesion and aggregation at sites of vascular injury is crucial for hemostasis but may lead to arterial occlusion in the setting of atherosclerosis and precipitate diseases such as myocardial infarction. A current hypothesis suggests that platelet glycoprotein (GP) Ib interaction with von Willebrand factor recruits flowing platelets to the injured vessel wall, where subendothelial fibrillar collagens support their firm adhesion and activation. However, so far this hypothesis has not been tested in vivo. Here, we demonstrate by intravital fluorescence microscopy of the mouse carotid artery that inhibition or absence of the major platelet collagen receptor, GPVI, abolishes platelet-vessel wall interactions after endothelial denudation. Unexpectedly, inhibition of GPVI by the monoclonal antibody JAQ1 reduced platelet tethering to the subendothelium by approximately 89%. In addition, stable arrest and aggregation of platelets was virtually abolished under these conditions. Using different models of arterial injury, the strict requirement for GPVI in these processes was confirmed in GPVI-deficient mice, where platelets also failed to adhere and aggregate on the damaged vessel wall. These findings reveal an unexpected role of GPVI in the initiation of platelet attachment at sites of vascular injury and unequivocally identify platelet-collagen interactions (via GPVI) as the major determinant of arterial thrombus formation.


Asunto(s)
Plaquetas/fisiología , Arterias Carótidas/patología , Glicoproteínas de Membrana Plaquetaria/metabolismo , Animales , Plaquetas/metabolismo , Adhesión Celular , Movimiento Celular , Colágeno/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Agregación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/deficiencia , Glicoproteínas de Membrana Plaquetaria/genética , Heridas y Lesiones/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...