Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Thorac Cardiovasc Surg ; 154(3): 978-988.e1, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28400112

RESUMO

BACKGROUND: Thrombosis persists as a leading cause of morbidity and mortality. Given that endothelial cells (ECs) play a central role in regulating thrombosis, understanding the molecular endothelial cues that regulate susceptibility or resistance to thrombosis have important translational implications. Accordingly, we evaluated the role of endothelial autophagy in the development of thrombosis. METHODS: We generated mice in which the essential autophagy-related 7 (ATG7) gene was conditionally deleted from ECs (EC-ATG7-/- mice). Three in vivo models of thrombosis were used, and mechanistic studies were conducted with cultured human umbilical vein endothelial cells (HUVECs). RESULTS: We silenced ATG7 in HUVECs and observed >60% decreases in tumor necrosis factor (TNF)-α-induced tissue factor (TF) transcript levels, protein expression, and activity. TF mRNA levels in the carotid arteries of EC-ATG7-/- mice subjected to the prothrombotic stimulus FeCl3 were lower than those in the similarly treated wild-type (WT) littermate group. Compared with WT mice, EC-ATG7-/- mice exhibited prolonged time to carotid (2-fold greater) and mesenteric (1.3-fold greater) artery occlusion following FeCl3 injury. The thrombi generated in laser-injured cremasteric arterioles were smaller in EC-ATG7-/- mice compared with WT mice, and took 2.3-fold longer to appear. CONCLUSIONS: Taken together, these results provide definitive evidence that loss of endothelial ATG7 attenuates thrombosis and reduces the expression of TF. Our findings demonstrate that endothelial ATG7, and thus autophagy, is a critical and previously unrecognized target for modulating the susceptibility to thrombosis.


Assuntos
Proteína 7 Relacionada à Autofagia , Células Endoteliais/metabolismo , Células Endoteliais da Veia Umbilical Humana/metabolismo , Trombose/metabolismo , Animais , Artérias Carótidas/metabolismo , Células Endoteliais/citologia , Predisposição Genética para Doença , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Artérias Mesentéricas/metabolismo , Camundongos Knockout , RNA Mensageiro/metabolismo , Trombose/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
2.
BMC Cardiovasc Disord ; 15: 130, 2015 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-26481314

RESUMO

Hemostasis encompasses a set of tightly regulated processes that govern blood clotting, platelet activation, and vascular repair. Upon vascular injury, the hemostatic system initiates a series of vascular events and activates extravascular receptors that act in concert to seal off the damage. Blood clotting is subsequently attenuated by a plethora of inhibitors that prevent excessive clot formation and eventual thrombosis. The endothelium which resides at the interface between the blood and surrounding tissues, serves an integral role in the hemostatic system. Depending on specific tissue needs and local stresses, endothelial cells are capable of evoking either antithrombotic or prothrombotic events. Healthy endothelial cells express antiplatelet and anticoagulant agents that prevent platelet aggregation and fibrin formation, respectively. In the face of endothelial dysfunction, endothelial cells trigger fibrin formation, as well as platelet adhesion and aggregation. Finally, endothelial cells release pro-fibrinolytic agents that initiate fibrinolysis to degrade the clot. Taken together, a functional endothelium is essential to maintain hemostasis and prevent thrombosis. Thus, a greater understanding into the role of the endothelium can provide new avenues for exploration and novel therapies for the management of thromboembolisms.


Assuntos
Células Endoteliais/fisiologia , Trombose/fisiopatologia , Coagulação Sanguínea/fisiologia , Hemostasia/fisiologia , Humanos , Neovascularização Fisiológica , Adesividade Plaquetária , Agregação Plaquetária
3.
Blood ; 123(13): 2102-7, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24501216

RESUMO

Central venous catheter thrombosis can cause venous obstruction and pulmonary embolism. To determine the extent to which catheter thrombosis is triggered by the contact or extrinsic pathway of coagulation, we used antisense oligonucleotides (ASOs) to selectively knock down factor (f)XII, fXI, or high-molecular-weight kininogen (HK), key components of the contact pathway, or fVII, which is essential for the extrinsic pathway. Knockdown of contact pathway components prolonged the activated partial thromboplastin time and decreased target protein activity levels by over 90%, whereas fVII knockdown prolonged the prothrombin time and reduced fVII activity to a similar extent. Using a rabbit model of catheter thrombosis, catheters implanted in the jugular vein were assessed daily until they occluded, up to a maximum of 35 days. Compared with control, fXII and fXI ASO treatment prolonged the time to catheter occlusion by 2.2- and 2.3-fold, respectively. In contrast, both HK and fVII knockdown did not significantly prolong the time to occlusion, and dual treatment with fVII- and fXI-directed ASOs produced a time to occlusion similar to that with the fXI ASO alone. These findings suggest that catheter thrombosis is triggered via the contact pathway and identify fXII and fXI as potential targets to attenuate this complication.


Assuntos
Catéteres/efeitos adversos , Fator XII/genética , Fator XI/genética , Oligonucleotídeos Antissenso/farmacologia , Interferência de RNA/fisiologia , Trombose/prevenção & controle , Animais , Obstrução do Cateter , Modelos Animais de Doenças , Fator XI/antagonistas & inibidores , Fator XII/antagonistas & inibidores , Regulação da Expressão Gênica/efeitos dos fármacos , Terapia Genética/métodos , Masculino , Coelhos , Especificidade por Substrato/efeitos dos fármacos , Especificidade por Substrato/genética , Trombose/genética
4.
Acta Biomater ; 8(11): 4092-100, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22824529

RESUMO

Catheters initiate coagulation by activating factor (f) XII, which can lead to catheter thrombosis. Fondaparinux, which only targets activated fX (fXa), is associated with more catheter thrombosis than heparin, which targets fXa and thrombin. To render catheters less thrombogenic and fondaparinux more effective, we examined whether coating catheters with corn trypsin inhibitor (CTI), which blocks fXIIa, attenuates catheter-induced clotting and promotes fondaparinux activity. Compared with unmodified catheters, CTI-coated catheters demonstrated (a) decreased adsorption of fibrinogen and fXII, (b) greater inhibition of fXIIa generated by catheter-induced autoactivation, (c) attenuated fXIIa-mediated activation of fXI and (d) longer plasma clotting times in the absence or presence of fondaparinux. In an accelerated catheter thrombosis model in rabbits, (a) the time to catheter occlusion was longer with CTI-coated catheters than with unmodified catheters and (b) an intravenous dose of fondaparinux that had no effect on the time to occlusion of unmodified catheters extended the time to occlusion of CTI-coated catheters. These findings support the concept that the prothrombotic activity of catheters reflects their capacity to activate fXII and identify CTI immobilization as a novel approach for rendering catheters and other blood-contacting medical devices less thrombogenic.


Assuntos
Catéteres/efeitos adversos , Materiais Revestidos Biocompatíveis/farmacologia , Proteínas de Plantas/farmacologia , Trombose/prevenção & controle , Adsorção , Animais , Coagulação Sanguínea/efeitos dos fármacos , Fator XII/metabolismo , Fondaparinux , Humanos , Intervenção Coronária Percutânea , Espectroscopia Fotoeletrônica , Polissacarídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Coelhos , Propriedades de Superfície/efeitos dos fármacos , Água/química
5.
J Biomed Mater Res A ; 100(4): 856-62, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22241533

RESUMO

The objective of this study was to investigate the bioactivity and protein-resistant properties of dual functioning surfaces modified with PEG for protein resistance and corn trypsin inhibitor (CTI) for anticoagulant effect. Surfaces on gold substrate were prepared with varying ratios of free PEG to CTI-conjugated PEG. Two methods designated, respectively, "sequential" and "direct" were used. For sequential surfaces, PEG was first immobilized on gold and the surfaces were incubated with CTI at varying concentration. For direct surfaces, a PEG-CTI conjugate was synthesized and gold surfaces were modified using solutions of the conjugate of varying concentration. The CTI density on these surfaces was measured using radiolabeled CTI. Water contact angles were measured and the thickness of PEG-CTI layers was determined by ellipsometry. Fibrinogen adsorption from buffer and human plasma, and adsorption from binary solutions of fibrinogen and α-lactalbumin were investigated using radiolabeling methods. Bioactivity of the surfaces was evaluated via their effects on FXIIa inhibition and plasma clotting time. It was found that as the ratio of CTI-conjugated PEG to free PEG increased, bioactivity increased but protein resistance was relatively constant. It is concluded that on these surfaces conjugation of PEG to CTI does not greatly compromise the protein resistance of the PEG but results in improved interactions between the CTI and the "target" protein FXIIa. At the same CTI density, sequential surfaces were more effective in terms of inhibiting FXIIa and prolonging clotting time.


Assuntos
Anticoagulantes/química , Proteínas de Plantas/química , Polietilenoglicóis/química , Proteínas/química
6.
J Biomater Sci Polym Ed ; 23(15): 1981-93, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-21986216

RESUMO

In previous work using gold as a model substrate, we showed that modification of surfaces with poly(ethylene glycol) (PEG) and corn trypsin inhibitor (CTI) rendered them protein resistant and inhibitory against activated factor XII. Sequential attachment of PEG followed by CTI gave superior performance compared to direct attachment of a preformed PEG-CTI conjugate. In the present work, a sequential method was used to attach PEG and CTI to a polyurethane (PU) substrate to develop a material with applicability for blood-contacting medical devices. Controls included surfaces modified only with PEG and only with CTI. Surfaces were characterized by water contact angle and X-ray photoelectron spectroscopy. The surface density of CTI was in the range of a monolayer and was higher on the PU substrate than on gold reported previously. Biointeractions were investigated by measuring fibrinogen adsorption from buffer and plasma, factor XIIa inhibition and plasma clotting time. Both the PU-PEG surfaces and the PU-PEG-CTI surfaces showed low fibrinogen adsorption from buffer and plasma, indicating that PEG retained its protein resistance when conjugated to CTI. Although the CTI density was lower on PU-PEG-CTI than on PU modified only with CTI, PU-PEG-CTI exhibited greater factor XIIa inhibition and a longer plasma clotting time, suggesting that PEG facilitates the interaction of CTI with factor XIIa. Thus sequential attachment of PEG and CTI may be a useful approach to improve the thromboresistance of PU surfaces.


Assuntos
Proteínas Sanguíneas/administração & dosagem , Materiais Revestidos Biocompatíveis/administração & dosagem , Proteínas de Plantas/administração & dosagem , Polietilenoglicóis/administração & dosagem , Poliuretanos/administração & dosagem , Coagulação Sanguínea/efeitos dos fármacos , Coagulação Sanguínea/fisiologia , Proteínas Sanguíneas/química , Materiais Revestidos Biocompatíveis/síntese química , Materiais Revestidos Biocompatíveis/química , Fator XIIa/química , Fator XIIa/metabolismo , Fibrinogênio/química , Fibrinogênio/metabolismo , Humanos , Espectroscopia Fotoeletrônica , Proteínas de Plantas/química , Polietilenoglicóis/química , Poliuretanos/química , Propriedades de Superfície
7.
Blood ; 118(25): 6667-74, 2011 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21937693

RESUMO

In patients undergoing percutaneous coronary intervention, catheter thrombosis is more frequent with fondaparinux than heparin. This study was undertaken to identify the responsible mechanism and to develop strategies for its prevention. Percutaneous coronary intervention catheter segments shortened plasma clotting times from 971 ± 92 to 352 ± 22 seconds. This activity is factor XII (fXII) dependent because it was attenuated with corn trypsin inhibitor and was abolished in fXII-deficient plasma. Heparin and enoxaparin blocked catheter-induced clotting at 0.5 and 2 anti-Xa U/mL, respectively, whereas fondaparinux had no effect. Addition of fondaparinux to bivalirudin or low-dose heparin attenuated catheter-induced clotting more than either agent alone. In a rabbit model of catheter thrombosis, a 70 anti-Xa U/kg intravenous bolus of heparin or enoxaparin prolonged the time to catheter occlusion by 4.6- and 2.5-fold, respectively, compared with saline, whereas the same dose of fondaparinux had no effect. Although 15 anti-Xa U/kg heparin had no effect on its own, when given in conjunction with 70 anti-Xa U/kg fondaparinux, the time to catheter occlusion was prolonged 2.9-fold. These findings indicate that (1) catheters are prothrombotic because they trigger fXII activation, and (2) fondaparinux does not prevent catheter-induced clotting unless supplemented with low-dose heparin or bivalirudin.


Assuntos
Enoxaparina/farmacologia , Heparina/farmacologia , Polissacarídeos/farmacologia , Trombose/prevenção & controle , Animais , Anticoagulantes/farmacologia , Coagulação Sanguínea/efeitos dos fármacos , Testes de Coagulação Sanguínea , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Catéteres/efeitos adversos , Modelos Animais de Doenças , Sinergismo Farmacológico , Fator XII/metabolismo , Fondaparinux , Humanos , Masculino , Coelhos , Trombose/sangue , Trombose/etiologia
8.
Acta Biomater ; 7(12): 4177-86, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21827874

RESUMO

Blood contacting surfaces bind plasma proteins and trigger coagulation by activating factor XII (FXII). The objective of this work was to develop blood contacting surfaces having the dual properties of protein resistance and inhibition of coagulation. Gold was used as a model substrate because it is amenable to facile modification using gold-thiol chemistry and to detailed surface characterization. The gold was modified with both polyethylene glycol (PEG) and corn trypsin inhibitor (CTI), a potent and specific inhibitor of activated FXII (FXIIa). Two methods of surface modification were developed; sequential and direct. In the sequential method PEG was first chemisorbed on gold; CTI was then attached to the PEG. In the direct method a conjugate of PEG and CTI was first prepared; the conjugate was then immobilized on gold. The surfaces were characterized by water contact angle and XPS. Biointeractions with the modified surfaces were assessed by measuring fibrinogen adsorption from buffer and plasma and by immunoblot analysis of eluted proteins after plasma exposure. Inhibition of FXIIa, autoactivation of FXII, and clotting times of plasma in contact with the surfaces were also measured. Both the sequential and direct surfaces showed reduced protein adsorption, increased FXIIa inhibition and longer clotting times compared with controls. Although the CTI density was lower on surfaces prepared using the sequential method, surfaces so prepared exhibited greater CTI activity than those generated by the direct method. It is concluded that the activity of immobilized PEG-CTI depends on the method of attachment and that immobilized CTI may be useful in rendering biomaterials more blood compatible.


Assuntos
Proteínas de Plantas/química , Polietilenoglicóis/química , Propriedades de Superfície , Zea mays/química , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
9.
Blood ; 117(15): 4134-41, 2011 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-21304106

RESUMO

Histidine-rich glycoprotein (HRG) circulates in plasma at a concentration of 2µM and binds plasminogen, fibrinogen, and thrombospondin. Despite these interactions, the physiologic role of HRG is unknown. Previous studies have shown that mice and humans deficient in HRG have shortened plasma clotting times. To better understand this phenomenon, we examined the effect of HRG on clotting tests. HRG prolongs the activated partial thromboplastin time in a concentration-dependent fashion but has no effect on tissue factor-induced clotting, localizing its effect to the contact pathway. Plasma immunodepleted of HRG exhibits a shortened activated partial thromboplastin time that is restored to baseline with HRG replenishment. To explore how HRG affects the contact pathway, we examined its binding to factors XII, XIIa, XI, and XIa. HRG binds factor XIIa with high affinity, an interaction that is enhanced in the presence of Zn²(+), but does not bind factors XII, XI, or XIa. In addition, HRG inhibits autoactivation of factor XII and factor XIIa-mediated activation of factor XI. These results suggest that, by binding to factor XIIa, HRG modulates the intrinsic pathway of coagulation, particularly in the vicinity of a thrombus where platelet release of HRG and Zn²(+) will promote this interaction.


Assuntos
Coagulação Sanguínea/fisiologia , Fator XIIa/metabolismo , Proteínas/metabolismo , Trombose/metabolismo , Testes de Coagulação Sanguínea , Fator XI/metabolismo , Fator XII/metabolismo , Fator XIa/metabolismo , Fibrinogênio/metabolismo , Humanos , Calicreínas/metabolismo , Pré-Calicreína/metabolismo , Zinco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...