Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Arterioscler Thromb Vasc Biol ; 35(8): 1798-804, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26069236

RESUMEN

OBJECTIVE: Clinical evidence suggests an association between oxidative stress and vascular disease, and in vitro studies have demonstrated that reactive oxygen species can have prothrombotic effects on vascular and blood cells. It remains unclear, however, whether elevated levels of reactive oxygen species accelerate susceptibility to experimental thrombosis in vivo. APPROACH AND RESULTS: Using a murine model with genetic deficiency in superoxide dismutase-1 (SOD1), we measured susceptibility to carotid artery thrombosis in response to photochemical injury. We found that SOD1-deficient (Sod1(-/-)) mice formed stable arterial occlusions significantly faster than wild-type (Sod1(+/+)) mice (P<0.05). Sod1(-/-) mice also developed significantly larger venous thrombi than Sod1(+/+) mice after inferior vena cava ligation (P<0.05). Activation of protein C by thrombin in lung was diminished in Sod1(-/-) mice (P<0.05 versus Sod1(+/+) mice), and generation of activated protein C in response to infusion of thrombin in vivo was decreased in Sod1(-/-) mice (P<0.05 versus Sod1(+/+) mice). SOD1 deficiency had no effect on the expression of thrombomodulin, endothelial protein C receptor, or tissue factor in lung or levels of protein C in plasma. Exposure of human thrombomodulin to superoxide in vitro caused oxidation of multiple methionine residues, including critical methionine 388, and a 40% decrease in thrombomodulin-dependent activation of protein C (P<0.05). SOD and catalase protected against superoxide-induced methionine oxidation and restored protein C activation in vitro (P<0.05). CONCLUSIONS: SOD prevents thrombomodulin methionine oxidation, promotes protein C activation, and protects against arterial and venous thrombosis in mice.


Asunto(s)
Traumatismos de las Arterias Carótidas/enzimología , Proteína C/metabolismo , Superóxido Dismutasa/deficiencia , Trombosis/enzimología , Trombosis de la Vena/enzimología , Animales , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Arteria Carótida Común/enzimología , Arteria Carótida Común/patología , Catalasa/metabolismo , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones de la Cepa 129 , Ratones Transgénicos , Oxidación-Reducción , Estrés Oxidativo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1 , Superóxidos/metabolismo , Trombomodulina/metabolismo , Trombosis/genética , Trombosis/patología , Vena Cava Inferior/enzimología , Vena Cava Inferior/patología , Trombosis de la Vena/patología
2.
Arterioscler Thromb Vasc Biol ; 35(4): 838-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25675995

RESUMEN

OBJECTIVE: We tested the hypothesis that endothelial peroxisome proliferator-activated receptor-γ protects against vascular thrombosis using a transgenic mouse model expressing a peroxisome proliferator-activated receptor-γ mutant (E-V290M) selectively in endothelium. APPROACH AND RESULTS: The time to occlusive thrombosis of the carotid artery was significantly shortened in E-V290M mice compared with nontransgenic littermates after either chemical injury with ferric chloride (5.1 ± 0.2 versus 10.1 ± 3.3 minutes; P=0.01) or photochemical injury with rose bengal (48 ± 9 versus 74 ± 9 minutes; P=0.04). Gene set enrichment analysis demonstrated the upregulation of NF-κB target genes, including P-selectin, in aortic endothelial cells from E-V290M mice (P<0.001). Plasma P-selectin and carotid artery P-selectin mRNA were elevated in E-V290M mice (P<0.05). P-selectin-dependent leukocyte rolling on mesenteric venules was increased in E-V290M mice compared with nontransgenic mice (53 ± 8 versus 25 ± 7 per minute; P=0.02). The shortened time to arterial occlusion in E-V290M mice was reversed by administration of P-selectin-blocking antibodies or neutrophil-depleting antibodies (P=0.04 and P=0.02, respectively) before photochemical injury. CONCLUSIONS: Endothelial peroxisome proliferator-activated receptor-γ protects against thrombosis through a mechanism that involves downregulation of P-selectin expression and diminished P-selectin-mediated leukocyte-endothelial interactions.


Asunto(s)
Enfermedades de las Arterias Carótidas/prevención & control , Células Endoteliales/metabolismo , Selectina-P/metabolismo , PPAR gamma/metabolismo , Trombosis/prevención & control , Trombosis de la Vena/prevención & control , Animales , Anticuerpos/farmacología , Enfermedades de las Arterias Carótidas/genética , Enfermedades de las Arterias Carótidas/inmunología , Enfermedades de las Arterias Carótidas/metabolismo , Enfermedades de las Arterias Carótidas/patología , Arteria Carótida Común/metabolismo , Arteria Carótida Común/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/patología , Humanos , Rodamiento de Leucocito , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Neutrófilos/inmunología , Neutrófilos/metabolismo , Selectina-P/antagonistas & inhibidores , Selectina-P/genética , Selectina-P/inmunología , PPAR gamma/genética , ARN Mensajero/metabolismo , Trombosis/genética , Trombosis/inmunología , Trombosis/metabolismo , Trombosis/patología , Factores de Tiempo , Vena Cava Inferior/metabolismo , Vena Cava Inferior/patología , Trombosis de la Vena/genética , Trombosis de la Vena/inmunología , Trombosis de la Vena/metabolismo , Trombosis de la Vena/patología
3.
Arterioscler Thromb Vasc Biol ; 35(12): 2594-604, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26449752

RESUMEN

OBJECTIVE: Emerging evidence suggests that methionine oxidation can directly affect protein function and may be linked to cardiovascular disease. The objective of this study was to define the role of the methionine sulfoxide reductase A (MsrA) in models of vascular disease and identify its signaling pathways. APPROACH AND RESULTS: MsrA was readily identified in all layers of the vascular wall in human and murine arteries. Deletion of the MsrA gene did not affect atherosclerotic lesion area in apolipoprotein E-deficient mice and had no significant effect on susceptibility to experimental thrombosis after photochemical injury. In contrast, the neointimal area after vascular injury caused by complete ligation of the common carotid artery was significantly greater in MsrA-deficient than in control mice. In aortic vascular smooth muscle cells lacking MsrA, cell proliferation was significantly increased because of accelerated G1/S transition. In parallel, cyclin D1 protein and cdk4/cyclin D1 complex formation and activity were increased in MsrA-deficient vascular smooth muscle cell, leading to enhanced retinoblastoma protein phosphorylation and transcription of E2F. Finally, MsrA-deficient vascular smooth muscle cell exhibited greater activation of extracellular signal-regulated kinase 1/2 that was caused by increased activity of the Ras/Raf/mitogen-activated protein kinase signaling pathway. CONCLUSIONS: Our findings implicate MsrA as a negative regulator of vascular smooth muscle cell proliferation and neointimal hyperplasia after vascular injury through control of the Ras/Raf/mitogen-activated protein kinase kinase/extracellular signal-regulated kinase 1/2 signaling pathway.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Traumatismos de las Arterias Carótidas/enzimología , Eliminación de Gen , Metionina Sulfóxido Reductasas/deficiencia , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neointima , Transducción de Señal , Trombosis/enzimología , Animales , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/patología , Arterias Carótidas/enzimología , Arterias Carótidas/patología , Traumatismos de las Arterias Carótidas/genética , Traumatismos de las Arterias Carótidas/patología , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Movimiento Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Hiperplasia , Masculino , Metionina Sulfóxido Reductasas/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Trombosis/sangre , Trombosis/genética , Factores de Tiempo , Quinasas raf/metabolismo , Proteínas ras/metabolismo
4.
Stroke ; 46(6): 1651-6, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25922506

RESUMEN

BACKGROUND AND PURPOSE: Cerebral aneurysm (CA) affects 3% of the population and is associated with hemodynamic stress and inflammation. Myeloperoxidase, a major oxidative enzyme associated with inflammation, is increased in patients with CA, but whether myeloperoxidase contributes to CA is not known. We tested the hypotheses that myeloperoxidase is increased within human CA and is critical for formation and rupture of CA in mice. METHODS: Blood was drawn from the lumen of CAs and femoral arteries of 25 patients who underwent endovascular coiling of CA, and plasma myeloperoxidase concentrations were measured with ELISA. Effects of endogenous myeloperoxidase on CA formation and rupture were studied in myeloperoxidase knockout mice and wild-type (WT) mice using an angiotensin II-elastase induction model of CA. In addition, effects of myeloperoxidase on inflammatory gene expression in endothelial cells were analyzed. RESULTS: Plasma concentrations of myeloperoxidase were 2.7-fold higher within CA than in femoral arterial blood in patients with CA. myeloperoxidase-positive cells were increased in aneurysm tissue compared with superficial temporal artery of patients with CA. Incidence of aneurysms and subarachnoid hemorrhage was significantly lower in myeloperoxidase knockout than in WT mice. In cerebral arteries, proinflammatory molecules, including tumor necrosis factor-α, cyclooxygenase-2 (COX2), chemokine (C-X-C motif) ligand 1 (CXCL1), chemokine (C motif) ligand (XCL1), matrix metalloproteinase (MMP) 8, cluster of differentiation 68 (CD68), and matrix metalloproteinase 13, and leukocytes were increased, and α-smooth muscle actin was decreased, in WT but not in myeloperoxidase knockout mice after induction of CA. Myeloperoxidase per se increased expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 in endothelial cells. CONCLUSIONS: These findings suggest that myeloperoxidase may contribute importantly to formation and rupture of CA.


Asunto(s)
Aneurisma Roto/sangre , Aneurisma Intracraneal/sangre , Peroxidasa/sangre , Aneurisma Roto/inducido químicamente , Aneurisma Roto/genética , Aneurisma Roto/patología , Angiotensina II/efectos adversos , Angiotensina II/farmacología , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Mediadores de Inflamación/sangre , Molécula 1 de Adhesión Intercelular/sangre , Molécula 1 de Adhesión Intercelular/genética , Aneurisma Intracraneal/inducido químicamente , Aneurisma Intracraneal/genética , Aneurisma Intracraneal/patología , Recuento de Leucocitos , Masculino , Ratones , Ratones Noqueados , Elastasa Pancreática/toxicidad , Peroxidasa/genética , Molécula 1 de Adhesión Celular Vascular/sangre , Molécula 1 de Adhesión Celular Vascular/genética , Vasoconstrictores/efectos adversos , Vasoconstrictores/farmacología
5.
Circulation ; 127(12): 1308-16, 2013 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-23426106

RESUMEN

BACKGROUND: The incidence of thrombotic events increases during aging, but the mechanisms are not well understood. To investigate the prothrombotic role of oxidative stress during aging, we tested the hypothesis that aged mice overexpressing the antioxidant enzyme glutathione peroxidase-1 (Gpx1) are protected from experimental thrombosis. METHODS AND RESULTS: Susceptibility to carotid artery thrombosis was first examined in wild-type C57BL/6J mice. After photochemical injury of the carotid artery, the time to stable occlusion was significantly shorter in 12- and 18-month-old mice compared with 4-month-old mice (P<0.01). Unlike wild-type mice, transgenic mice overexpressing Gpx1 (Gpx1 Tg) did not exhibit shortened times to occlusion of the carotid artery at 12 or 18 months of age. Wild-type mice also exhibited increased susceptibility to venous thrombosis after inferior vena cava ligation at 12 or 18 months of age (P<0.05 versus 4 months of age). Gpx1 Tg mice were protected from this aging-related enhanced susceptibility to venous thrombosis. Age-dependent platelet hyperactivation, evidenced by increased hydrogen peroxide, fibrinogen binding, and activation of fibrinogen receptor αIIbß3, was observed in thrombin-activated platelets from wild-type but not Gpx1 Tg mice (P<0.05). Enhanced platelet activation responses in aged mice were also prevented by polyethylene glycol-catalase or apocynin, an inhibitor of NADPH oxidase. Aged mice displayed increased intraplatelet expression of p47(phox) and superoxide dismutase-1, suggesting a mechanistic pathway for increased hydrogen peroxide generation. CONCLUSIONS: Our findings demonstrate that hydrogen peroxide is a key mediator of platelet hyperactivity and enhanced thrombotic susceptibility in aged mice.


Asunto(s)
Envejecimiento/metabolismo , Plaquetas/metabolismo , Peróxido de Hidrógeno/metabolismo , Activación Plaquetaria/fisiología , Trombosis/epidemiología , Trombosis/metabolismo , Acetofenonas/farmacología , Animales , Catalasa/farmacología , Femenino , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Animales , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Estrés Oxidativo/fisiología , Activación Plaquetaria/efectos de los fármacos , Polietilenglicoles/farmacología , Factores de Riesgo , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1 , Trombosis/fisiopatología , Glutatión Peroxidasa GPX1
6.
J Thromb Haemost ; 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38815756

RESUMEN

BACKGROUND: COVID-19 can cause profound inflammation and coagulopathy, and while many mechanisms have been proposed, there is no known common pathway leading to a prothrombotic state. OBJECTIVES: From the beginning of the COVID-19 pandemic, elevated levels of extracellular histones have been found in plasma of patients infected with SARS-CoV-2. We hypothesized that platelet activation triggered by extracellular histones might represent a unifying mechanism leading to increased thrombin generation and thrombosis. METHODS: We utilized blood samples collected from an early clinical trial of hospitalized COVID-19 patients (NCT04360824) and recruited healthy subjects as controls. Using plasma samples, we measured the procoagulant and prothrombotic potential of circulating extracellular histones and extracellular vesicles (EVs). Platelet prothrombotic activity was assessed via thrombin generation potential and platelet thrombus growth. Circulating EVs were assessed for thrombin generation potential in vitro in plasma and enhancement of thrombotic susceptibility in vivo in mice. RESULTS: Compared with controls, COVID-19 patients had elevated plasma levels of citrullinated histone H3, cell-free DNA, nucleosomes, and EVs. Plasma from COVID-19 patients promoted platelet activation, platelet-dependent thrombin generation, thrombus growth under venous shear stress, and release of platelet-derived EVs. These prothrombotic effects of COVID-19 plasma were inhibited by an RNA aptamer that neutralizes both free and DNA-bound histones. EVs isolated from COVID-19 plasma enhanced thrombin generation in vitro and potentiated venous thrombosis in mice in vivo. CONCLUSION: We conclude that extracellular histones and procoagulant EVs drive the prothrombotic state in COVID-19 and that histone-targeted therapy may prove beneficial.

7.
Circ Res ; 106(3): 551-8, 2010 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-20019334

RESUMEN

RATIONALE: Hyperhomocysteinemia is a cardiovascular risk factor that is associated with elevation of the nitric oxide synthase inhibitor asymmetrical dimethylarginine (ADMA). OBJECTIVE: Using mice transgenic for overexpression of the ADMA-hydrolyzing enzyme dimethylarginine dimethylaminohydrolase-1 (DDAH1), we tested the hypothesis that overexpression of DDAH1 protects from adverse structural and functional changes in cerebral arterioles in hyperhomocysteinemia. METHODS AND RESULTS: Hyperhomocysteinemia was induced in DDAH1 transgenic (DDAH1 Tg) mice and wild-type littermates using a high methionine/low folate (HM/LF) diet. Plasma total homocysteine was elevated approximately 3-fold in both wild-type and DDAH1 Tg mice fed the HM/LF diet compared with the control diet (P<0.001). Plasma ADMA was approximately 40% lower in DDAH1 Tg mice compared with wild-type mice (P<0.001) irrespective of diet. Compared with the control diet, the HM/LF diet diminished endothelium-dependent dilation to 10 micromol/L acetylcholine in cerebral arterioles of both wild-type (12 + or - 2 versus 29 + or - 3%; P<0.001) and DDAH1 Tg (14 + or - 3 versus 28 + or - 2%; P<0.001) mice. Responses to 10 micromol/L papaverine, a direct smooth muscle dilator, were impaired with the HM/LF diet in wild-type mice (30 + or - 3 versus 45 + or - 5%; P<0.05) but not DDAH1 Tg mice (45 + or - 7 versus 48 + or - 6%). DDAH1 Tg mice also were protected from hypertrophy of cerebral arterioles (P<0.05) but not from accelerated carotid artery thrombosis induced by the HM/LF diet. CONCLUSIONS: Overexpression of DDAH1 protects from hyperhomocysteinemia-induced alterations in cerebral arteriolar structure and vascular muscle function.


Asunto(s)
Amidohidrolasas/fisiología , Arteriolas/patología , Enfermedades Arteriales Cerebrales/prevención & control , Hiperhomocisteinemia/prevención & control , Acetilcolina/farmacología , Amidohidrolasas/genética , Animales , Arginina/análogos & derivados , Arginina/sangre , Trombosis de las Arterias Carótidas/etiología , Enfermedades Arteriales Cerebrales/etiología , Dieta/efectos adversos , Deficiencia de Ácido Fólico/complicaciones , Humanos , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/patología , Hipertrofia , Metionina/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Papaverina/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
8.
Arterioscler Thromb Vasc Biol ; 31(11): 2509-17, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21885846

RESUMEN

OBJECTIVE: We sought to develop a murine model to examine the antithrombotic and antiinflammatory functions of human thrombomodulin in vivo. METHODS AND RESULTS: Knock-in mice that express human thrombomodulin from the murine thrombomodulin gene locus were generated. Compared with wild-type mice, human thrombomodulin knock-in mice exhibited decreased protein C activation in the aorta (P<0.01) and lung (P<0.001). Activation of endogenous protein C following infusion of thrombin was decreased by 90% in knock-in mice compared with wild-type mice (P<0.05). Carotid artery thrombosis induced by photochemical injury occurred more rapidly in knock-in mice (12±3 minutes) than in wild-type mice (31±6 minutes; P<0.05). No differences in serum cytokine levels were detected between knock-in and wild-type mice after injection of endotoxin. When crossed with apolipoprotein E-deficient mice and fed a Western diet, knock-in mice had a further decrease in protein C activation but did not exhibit increased atherosclerosis. CONCLUSION: Expression of human thrombomodulin in place of murine thrombomodulin produces viable mice with a prothrombotic phenotype but unaltered responses to systemic inflammatory or atherogenic stimuli. This humanized animal model will be useful for investigating the function of human thrombomodulin under pathophysiological conditions in vivo.


Asunto(s)
Aterosclerosis/fisiopatología , Modelos Animales de Enfermedad , Trombomodulina/genética , Trombomodulina/fisiología , Trombosis/fisiopatología , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Aterosclerosis/metabolismo , Citocinas/sangre , Citocinas/efectos de los fármacos , Endotoxinas/farmacología , Femenino , Técnicas de Sustitución del Gen , Humanos , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa C/metabolismo , Especificidad de la Especie , Trombosis/metabolismo
9.
Arterioscler Thromb Vasc Biol ; 27(1): 233-40, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17082485

RESUMEN

OBJECTIVE: We tested the hypothesis that hyperhomocysteinemia and hypercholesterolemia promote arterial thrombosis in mice. METHODS AND RESULTS: Male apolipoprotein E (Apoe)-deficient mice were fed one of four diets: control, hyperhomocysteinemic (HH), high fat (HF), or high fat/hyperhomocysteinemic (HF/HH). Total cholesterol was elevated 2-fold with the HF or HF/HH diets compared with the control or HH diets (P<0.001). Plasma total homocysteine (tHcy) was elevated (12 to 15 micromol/L) with the HH or HF/HH diets compared with the control or HF diets (4 to 6 micromol/L; P<0.001). Aortic sinus lesion area correlated strongly with total cholesterol (P<0.001) but was independent of tHcy. At 12 weeks of age, the time to thrombotic occlusion of the carotid artery after photochemical injury was >50% shorter in mice fed the HF diets, with or without hyperhomocysteinemia, compared with the control diet (P<0.05). At 24 weeks of age, carotid artery thrombosis was also accelerated in mice fed the HH diet (P<0.05). Endothelium-dependent nitric oxide-mediated relaxation of carotid artery rings was impaired in mice fed the HF, HH, or HF/HH diets compared with the control diet (P<0.05). CONCLUSIONS: Hyperhomocysteinemia and hypercholesterolemia, alone or in combination, produce endothelial dysfunction and increased susceptibility to thrombosis in Apoe-deficient mice.


Asunto(s)
Apolipoproteínas E/fisiología , Hipercolesterolemia/fisiopatología , Hiperhomocisteinemia/fisiopatología , Trombosis/fisiopatología , Animales , Apolipoproteínas E/genética , Peso Corporal/fisiología , Colesterol/sangre , Susceptibilidad a Enfermedades/fisiopatología , Endotelio Vascular/fisiopatología , Regulación de la Expresión Génica/fisiología , Homocisteína/sangre , Hipercolesterolemia/complicaciones , Hipercolesterolemia/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/metabolismo , Masculino , Metionina/sangre , Ratones , Ratones Noqueados , Activación Plaquetaria/fisiología , Trombosis/etiología , Trombosis/metabolismo
10.
JCI Insight ; 1(7)2016.
Artículo en Inglés | MEDLINE | ID: mdl-27294204

RESUMEN

Reperfusion injury can exacerbate tissue damage in ischemic stroke, but little is known about the mechanisms linking ROS to stroke severity. Here, we tested the hypothesis that protein methionine oxidation potentiates NF-κB activation and contributes to cerebral ischemia/reperfusion injury. We found that overexpression of methionine sulfoxide reductase A (MsrA), an antioxidant enzyme that reverses protein methionine oxidation, attenuated ROS-augmented NF-κB activation in endothelial cells, in part, by protecting against the oxidation of methionine residues in the regulatory domain of calcium/calmodulin-dependent protein kinase II (CaMKII). In a murine model, MsrA deficiency resulted in increased NF-κB activation and neutrophil infiltration, larger infarct volumes, and more severe neurological impairment after transient cerebral ischemia/reperfusion injury. This phenotype was prevented by inhibition of NF-κB or CaMKII. MsrA-deficient mice also exhibited enhanced leukocyte rolling and upregulation of E-selectin, an endothelial NF-κB-dependent adhesion molecule known to contribute to neurovascular inflammation in ischemic stroke. Finally, bone marrow transplantation experiments demonstrated that the neuroprotective effect was mediated by MsrA expressed in nonhematopoietic cells. These findings suggest that protein methionine oxidation in nonmyeloid cells is a key mechanism of postischemic oxidative injury mediated by NF-κB activation, leading to neutrophil recruitment and neurovascular inflammation in acute ischemic stroke.

11.
Hypertension ; 66(1): 211-20, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25916724

RESUMEN

Vascular inflammation plays a critical role in the pathogenesis of cerebral aneurysms. Peroxisome proliferator-activated receptor γ (PPARγ) protects against vascular inflammation and atherosclerosis, whereas dominant-negative mutations in PPARγ promote atherosclerosis and vascular dysfunction. We tested the role of PPARγ in aneurysm formation and rupture. Aneurysms were induced with a combination of systemic infusion of angiotensin-II and local injection of elastase in (1) mice that received the PPARγ antagonist GW9662 or the PPARγ agonist pioglitazone, (2) mice carrying dominant-negative PPARγ mutations in endothelial or smooth muscle cells, and (3) mice that received the Cullin inhibitor MLN4924. Incidence of aneurysm formation, rupture, and mortality was quantified. Cerebral arteries were analyzed for expression of Cullin3, Kelch-like ECH-associated protein 1, nuclear factor (erythroid-derived 2)-like 2, NAD(P)H dehydrogenase (quinone)1 (NQO1), and inflammatory marker mRNAs. Neither pioglitazone nor GW9662 altered the incidence of aneurysm formation. GW9662 significantly increased the incidence of aneurysm rupture, whereas pioglitazone tended to decrease the incidence of rupture. Dominant-negative endothelial-specific PPARγ did not alter the incidence of aneurysm formation or rupture. In contrast, dominant-negative smooth muscle-specific PPARγ resulted in an increase in aneurysm formation (P<0.05) and rupture (P=0.05). Dominant-negative smooth muscle-specific PPARγ, but not dominant-negative endothelial-specific PPARγ, resulted in significant decreases in expression of genes encoding Cullin3, Kelch-like ECH-associated protein 1, and nuclear factor (erythroid-derived 2)-like 2, along with significant increases in tumor necrosis factor-α, monocyte chemoattractant protein-1, chemokine (C-X-C motif) ligand 1, CD68, matrix metalloproteinase-3, -9, and -13. MLN4924 did not alter incidence of aneurysm formation, but increased the incidence of rupture (P<0.05). In summary, endogenous PPARγ, specifically smooth muscle PPARγ, plays an important role in protecting from formation and rupture of experimental cerebral aneurysms in mice.


Asunto(s)
Aneurisma Roto/fisiopatología , Aneurisma Intracraneal/fisiopatología , PPAR gamma/fisiología , Aneurisma Roto/genética , Angiotensina II/toxicidad , Anilidas/farmacología , Anilidas/toxicidad , Animales , Arterias Cerebrales/metabolismo , Endotelio Vascular/metabolismo , Regulación de la Expresión Génica/fisiología , Genes Dominantes , Hipertensión/inducido químicamente , Hipertensión/complicaciones , Mediadores de Inflamación/metabolismo , Aneurisma Intracraneal/inducido químicamente , Aneurisma Intracraneal/genética , Ratones , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Miocitos del Músculo Liso/metabolismo , Especificidad de Órganos , PPAR gamma/agonistas , PPAR gamma/antagonistas & inhibidores , PPAR gamma/deficiencia , PPAR gamma/genética , Elastasa Pancreática/toxicidad , Pioglitazona , Hemorragia Subaracnoidea/etiología , Hemorragia Subaracnoidea/prevención & control , Tiazolidinedionas/farmacología , Regulación hacia Arriba , Vasculitis/complicaciones , Vasculitis/genética , Vasculitis/metabolismo , Vasculitis/patología
13.
Blood ; 111(3): 1257-65, 2008 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17989312

RESUMEN

Many of the cellular responses that occur in activated platelets resemble events that take place following activation of cell-death pathways in nucleated cells. We tested the hypothesis that formation of the mitochondrial permeability transition pore (MPTP), a key signaling event during cell death, also plays a critical role in platelet activation. Stimulation of murine platelets with thrombin plus the glycoprotein VI agonist convulxin resulted in a rapid loss of mitochondrial transmembrane potential (Deltapsi(m)) in a subpopulation of activated platelets. In the absence of cyclophilin D (CypD), an essential regulator of MPTP formation, murine platelet activation responses were altered. CypD-deficient platelets exhibited defects in phosphatidylserine externalization, high-level surface fibrinogen retention, membrane vesiculation, and procoagulant activity. Also, in CypD-deficient platelet-rich plasma, clot retraction was altered. Stimulation with thrombin plus H(2)O(2), a known activator of MPTP formation, also increased high-level surface fibrinogen retention, phosphatidylserine externalization, and platelet procoagulant activity in a CypD-dependent manner. In a model of carotid artery photochemical injury, thrombosis was markedly accelerated in CypD-deficient mice. These results implicate CypD and the MPTP as critical regulators of platelet activation and suggest a novel CypD-dependent negative-feedback mechanism regulating arterial thrombosis.


Asunto(s)
Ciclofilinas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Activación Plaquetaria , Trombosis/metabolismo , Animales , Peptidil-Prolil Isomerasa F , Ciclofilinas/deficiencia , Ciclofilinas/genética , Peróxido de Hidrógeno/farmacología , Ionomicina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Poro de Transición de la Permeabilidad Mitocondrial , Activación Plaquetaria/efectos de los fármacos , Trombosis/genética
14.
Blood ; 108(7): 2237-43, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16804115

RESUMEN

Hyperhomocysteinemia is a risk factor for thrombosis, but the mechanisms are not well defined. We tested the hypothesis that hyperhomocysteinemia accelerates arterial thrombosis in mice. Mice heterozygous for a targeted disruption of the cystathionine beta-synthase gene (Cbs+/-) and wild-type littermates (Cbs+/+) were fed either a control diet or a high methionine/low folate (HM/LF) diet for 6 to 8 months to produce graded hyperhomocysteinemia. The time to occlusion of the carotid artery after photochemical injury was shortened by more than 50% in Cbs+/+ or Cbs+/- mice fed the HM/LF diet (P < .001 versus control diet). Carotid artery thrombosis was not accelerated in mice deficient in endothelial nitric oxide synthase (Nos3), which suggests that decreased endothelium-derived nitric oxide is not a sufficient mechanism for enhancement of thrombosis. Cbs+/+ and Cbs+/- mice fed the HM/LF diet had elevated levels of reactive oxygen species in the carotid artery, increased aortic expression of the NADPH oxidase catalytic subunit, Nox4, and decreased activation of anticoagulant protein C in the aorta (P < .05 versus control diet). We conclude that hyperhomocysteinemia enhances susceptibility to arterial thrombosis through a mechanism that is not caused by loss of endothelium-derived nitric oxide but may involve oxidative stress and impairment of the protein C anticoagulant pathway.


Asunto(s)
Arterias Carótidas/patología , Predisposición Genética a la Enfermedad , Hiperhomocisteinemia/diagnóstico , Hiperhomocisteinemia/genética , Trombosis/patología , Animales , Anticoagulantes/metabolismo , Cistationina betasintasa/genética , Hiperhomocisteinemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III , Especies Reactivas de Oxígeno
15.
Semin Vasc Med ; 5(2): 163-71, 2005 May.
Artículo en Inglés | MEDLINE | ID: mdl-16047268

RESUMEN

Hyperhomocysteinemia is a risk factor for cardiovascular disease and stroke. During the last decade, considerable progress in delineating the mechanisms that underlie the atherogenic effects of hyperhomocysteinemia has been achieved through the use of experimental animal models. Among the most informative animal models are those that use genetic and dietary approaches to produce hyperhomocysteinemia in mice. Recent findings demonstrate that hyperhomocysteinemia can accelerate the development of atherosclerosis in susceptible models such as the apolipoprotein E-deficient mouse. Hyperhomocysteinemia also is a potent inducer of endothelial dysfunction, particularly in small vessels such as cerebral arterioles. Mechanisms of endothelial dysfunction may include inhibition of endothelial nitric oxide synthase by its endogenous inhibitor, asymmetric dimethylarginine, and oxidative inactivation of nitric oxide mediated by upregulation of prooxidant enzymes and downregulation of antioxidant enzymes. There also is good evidence from animal models that hyperhomocysteinemia produces endoplasmic reticulum stress, which may contribute to atherosclerosis and endothelial dysfunction by activating signal transduction pathways leading to inflammation, oxidative stress, and apoptosis.


Asunto(s)
Arteriosclerosis/etiología , Homocisteína/sangre , Hiperhomocisteinemia/complicaciones , Animales , Arteriosclerosis/sangre , Arteriosclerosis/fisiopatología , Modelos Animales de Enfermedad , Endotelio Vascular/metabolismo , Endotelio Vascular/fisiopatología , Hiperhomocisteinemia/sangre , Hiperhomocisteinemia/inducido químicamente , Metionina/toxicidad , Ratones , Óxido Nítrico/metabolismo , Estrés Oxidativo , Factores de Riesgo
16.
Biochem Biophys Res Commun ; 293(1): 274-83, 2002 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-12054596

RESUMEN

Peroxisome proliferator activated receptor-gamma (PPAR-gamma) is abundantly expressed in atherosclerotic lesions and is implicated in atherogenesis. The existence of three splice variants, PPAR-gamma 1, PPAR-gamma 2, and PPAR-gamma 3 has been established. Using monocyte-derived macrophages from cynomolgus monkeys, we demonstrate here the identification of two new PPAR-gamma exons, exon C and exon D, which splice together with already established exons A1, A2, and B in the 5(') terminal region to generate four novel PPAR-gamma subtypes, PPAR-gamma 4, -gamma 5, -gamma 6, and -gamma 7. PPAR-gamma 4 and gamma 5 were detected only in macrophages whereas gamma 6 and gamma 7 were expressed both in macrophages and adipose tissues. None of these novel isoforms were detected in muscle, kidney, and spleen from monkeys. We found sequences identical to exons C and D in the human genome database. These and all PPAR-gamma exons known to date are encoded by a single gene, located from region 10498 K to 10384 K on human chromosome 3. We cloned and expressed PPAR-gamma 1, PPAR-gamma 4, and PPAR-gamma 5 proteins in yeast using the expression vector pPICZB. As expected, all recombinant proteins showed a molecular weight of approximately 50 kDa. We also investigated the effect of a high-fat diet on the level of macrophage PPAR-gamma expression in monkeys. RT-PCR showed a significant increase in total PPAR-gamma and ABCA1 mRNA levels in macrophages of fat-fed monkeys (n=7) compared to those maintained on a normal diet (n=2). However, none of the novel isoforms seemed to be induced by fat-feeding. We used tetracycline-responsive expression vectors to obtain moderate expression of PPAR-gamma 4 and -gamma 5 in CHO cells. In these cells, expression of PPAR-gamma 5 but not -gamma 4 repressed the expression of ABCA1. Neither isoform modulated the expression of lipoprotein lipase. Our results suggest that individual PPAR-gamma isoforms may be responsible for unique tissue-specific biological effects and that PPAR-gamma 4 and -gamma 5 may modulate macrophage function and atherogenesis.


Asunto(s)
Empalme Alternativo , Receptores Citoplasmáticos y Nucleares/genética , Factores de Transcripción/genética , Animales , Cartilla de ADN , Exones , Variación Genética , Haplorrinos , Humanos , Macaca fascicularis , Macaca mulatta , Ratones , Isoformas de Proteínas/genética , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA