Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Cereb Blood Flow Metab ; 39(11): 2233-2245, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-30073881

RESUMO

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is implicated in cardiovascular disease by modulating apoptosis and oxidative stress. We hypothesized that LOX-1 may be involved in pathophysiology of stroke by mediating ischaemia/reperfusion (I/R)-dependent cell death. Transient middle cerebral artery occlusion (tMCAO) was performed in wild-type (WT) mice, endothelial-specific LOX-1 transgenic mice (eLOX-1TG) and WT animals treated with LOX-1 silencing RNA (siRNA). In WT mice exposed to tMCAO, LOX-1 expression and function were increased in the MCA. Compared to WT animals, eLOX-1TG mice displayed increased stroke volumes and worsened outcome after I/R. Conversely, LOX-1-silencing decreased both stroke volume and neurological impairment. Similarly, in HBMVECs, hypoxia/reoxygenation increased LOX-1 expression, while LOX-1 overexpressing cells showed increased death following hypoxia reoxygenation. Increased caspase-3 activation was observed following LOX-1 overexpression both in vivo and in vitro, thus representing a likely mediator. Finally, monocytes from ischaemic stroke patients exhibited increased LOX-1 expression which also correlated with disease severity. Our data unequivocally demonstrate a key role for LOX-1 in determining outcome following I/R brain damage. Our findings could be corroborated in human brain endothelial cells and monocytes from patients, underscoring their translational relevance and suggesting siRNA-mediated LOX-1 knockdown as a novel therapeutic strategy for stroke patients.


Assuntos
Encéfalo/patologia , Traumatismo por Reperfusão/etiologia , Receptores Depuradores Classe E/fisiologia , Acidente Vascular Cerebral/etiologia , Animais , Apoptose , Lesões Encefálicas/etiologia , Células Cultivadas , Células Endoteliais/metabolismo , Humanos , Infarto da Artéria Cerebral Média , Camundongos , Camundongos Transgênicos , Monócitos/metabolismo , Estresse Oxidativo , RNA Interferente Pequeno/genética
2.
Cardiovasc Res ; 113(5): 498-507, 2017 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-28199510

RESUMO

AIMS: The lectin-like oxLDL receptor-1 (LOX-1) promotes endothelial uptake of oxidized low-density lipoprotein (oxLDL) and plays an important role in atherosclerosis and acute coronary syndromes (ACS). However, its role in arterial thrombus formation remains unknown. We investigated whether LOX-1 plays a role in arterial thrombus formation in vivo at different levels of oxLDL using endothelial-specific LOX-1 transgenic mice (LOX-1TG) and a photochemical injury thrombosis model of the carotid artery. METHODS AND RESULTS: In mice fed a normal chow diet, time to arterial occlusion was unexpectedly prolonged in LOX-1TG as compared to WT. In line with this, tissue factor (TF) expression and activity in carotid arteries of LOX-1TG mice were reduced by half. This effect was mediated by activation of octamer transcription factor 1 (Oct-1) leading to upregulation of the mammalian deacetylase silent information regulator-two 1 (SIRT1) via binding to its promoter and subsequent inhibition of NF-κB signaling. In contrast, intravenous injection of oxLDL as well as high cholesterol diet for 6 weeks led to a switch from the Oct-1/SIRT1 signal transduction pathway to the ERK1/2 pathway and in turn to an enhanced thrombotic response with shortened occlusion time. CONCLUSIONS: Thus, LOX-1 differentially regulates thrombus formation in vivo depending on the degree of activation by oxLDL. At low oxLDL levels LOX-1 activates the protective Oct-1/SIRT1 pathway, while at higher levels of the lipoprotein switches to the thrombogenic ERK1/2 pathway. These findings may be important for arterial thrombus formation in ACS and suggest that SIRT1 may represent a novel therapeutic target in this context.


Assuntos
Coagulação Sanguínea , Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/enzimologia , Lipoproteínas LDL/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator 1 de Transcrição de Octâmero/metabolismo , Receptores Depuradores Classe E/metabolismo , Sirtuína 1/metabolismo , Trombose/enzimologia , Animais , Sítios de Ligação , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/genética , Colesterol na Dieta , Modelos Animais de Doenças , Predisposição Genética para Doença , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , NF-kappa B/genética , NF-kappa B/metabolismo , Fator 1 de Transcrição de Octâmero/genética , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Receptores Depuradores Classe E/genética , Transdução de Sinais , Sirtuína 1/genética , Tromboplastina/metabolismo , Trombose/sangue , Trombose/genética , Trombose/prevenção & controle , Fatores de Tempo
3.
Am J Physiol Heart Circ Physiol ; 312(2): H232-H238, 2017 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-27836896

RESUMO

In Alzheimer's disease (AD), cerebral arteries, in contrast to cerebral microvessels, show both cerebral amyloid angiopathy (CAA) -dependent and -independent vessel wall pathology. However, it remains unclear whether CAA-independent vessel wall pathology affects arterial function, thereby chronically reducing cerebral perfusion, and, if so, which mechanisms mediate this effect. To this end, we assessed the ex vivo vascular function of the basilar artery and a similar-sized peripheral artery (femoral artery) in the Swedish-Arctic (SweArc) transgenic AD mouse model at different disease stages. Furthermore, we used quantitative immunohistochemistry to analyze CAA, endothelial morphology, and molecular pathways pertinent to vascular relaxation. We found that endothelium-dependent, but not smooth muscle-dependent, vasorelaxation was significantly impaired in basilar and femoral arteries of 15-mo-old SweArc mice compared with that of age-matched wild-type and 6-mo-old SweArc mice. This impairment was accompanied by significantly reduced levels of cyclic GMP, indicating a reduced nitric oxide (NO) bioavailability. However, no age- and genotype-related differences in oxidative stress as measured by lipid peroxidation were observed. Although parenchymal capillaries, arterioles, and arteries showed abundant CAA in the 15-mo-old SweArc mice, no CAA or changes in endothelial morphology were detected histologically in the basilar and femoral artery. Thus our results suggest that, in this AD mouse model, dysfunction of large intracranial, extracerebral arteries important for brain perfusion is mediated by reduced NO bioavailability rather than by CAA. This finding supports the growing body of evidence highlighting the therapeutic importance of targeting the cerebrovasculature in AD. NEW & NOTEWORTHY: We show that vasorelaxation of the basilar artery, a large intracranial, extracerebral artery important for cerebral perfusion, is impaired independent of cerebral amyloid angiopathy in a transgenic mouse model of Alzheimer's disease. Interestingly, this dysfunction is specifically endothelium related and is mediated by impaired nitric oxide-cyclic GMP bioavailability.


Assuntos
Envelhecimento/metabolismo , Doença de Alzheimer/metabolismo , Peptídeos beta-Amiloides/metabolismo , Artéria Basilar/metabolismo , Angiopatia Amiloide Cerebral/metabolismo , Óxido Nítrico/metabolismo , Envelhecimento/fisiologia , Doença de Alzheimer/patologia , Doença de Alzheimer/fisiopatologia , Animais , Artéria Basilar/patologia , Artéria Basilar/fisiopatologia , Disponibilidade Biológica , Angiopatia Amiloide Cerebral/patologia , Angiopatia Amiloide Cerebral/fisiopatologia , Artérias Cerebrais/metabolismo , Artérias Cerebrais/patologia , Artérias Cerebrais/fisiopatologia , Circulação Cerebrovascular/fisiologia , GMP Cíclico/metabolismo , Modelos Animais de Doenças , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Artéria Femoral/metabolismo , Artéria Femoral/patologia , Artéria Femoral/fisiopatologia , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Transgênicos , Vasodilatação/fisiologia
4.
Thromb Haemost ; 117(1): 83-89, 2017 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-27761579

RESUMO

Despite public awareness of its deleterious effects, smoking remains a major cause of death. Indeed, it is a risk factor for atherothrombotic complications and in line with this, the introduction of smoking ban in public areas reduced smoking-associated cardiovascular complications. Nonetheless, smoking remains a major concern, and molecular mechanisms by which it causes cardiovascular disease are not known. Peripheral blood monocytes from healthy smokers displayed increased JNK2 and tissue factor (TF) gene expression compared to non-smokers (n=15, p<0.05). Similarly, human aortic endothelial cells exposed to cigarette smoke total particulate matter (CS-TPM) revealed increased TF expression mediated by JNK2 (n=4; p<0.05). Wild-type and JNK2-/- mice were exposed to cigarette smoke for two weeks after which arterial thrombosis was investigated. Wild-type mice exposed to smoke displayed reduced time to thrombotic arterial occlusion (n=8; p<0.05) and increased tissue factor activity (n=7; p<0.05) as compared to wild-type controls (n=6), while JNK2-/-mice exposed to smoke maintained an unaltered thrombotic potential (n=8; p=NS) and tissue factor activity (n=8) comparable to that of JNK2-/- and wild-type controls (n=6; p=NS). Smoking caused an increased production of reactive oxygen species (ROS) in wild-type but not in JNK2-/- mice (n=7; p<0.05 for wild-type mice and n=5-6; p=NS for JNK2-/- mice). In conclusion, the MAP kinase JNK2 mediates cigarette smoke-induced TF activation, arterial thrombosis and ROS production. These results underscore a major role of JNK2 in smoke-mediated thrombus formation and may offer an attractive target to prevent smoke-related thrombosis in those subjects which do not manage quitting.


Assuntos
Arteriopatias Oclusivas/etiologia , Coagulação Sanguínea , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Fumaça/efeitos adversos , Fumar/efeitos adversos , Trombose/etiologia , Animais , Arteriopatias Oclusivas/sangue , Arteriopatias Oclusivas/enzimologia , Arteriopatias Oclusivas/genética , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/enzimologia , Lesões das Artérias Carótidas/genética , Células Cultivadas , Células Endoteliais/enzimologia , Feminino , Predisposição Genética para Doença , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/deficiência , Proteína Quinase 9 Ativada por Mitógeno/genética , Fenótipo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Fumar/sangue , Fumar/genética , Tromboplastina/genética , Tromboplastina/metabolismo , Trombose/sangue , Trombose/enzimologia , Trombose/genética
5.
Hypertension ; 64(2): 347-53, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24842918

RESUMO

Increased cyclic stretch to the vessel wall, as observed in hypertension, leads to endothelial dysfunction through increased free radical production and reduced nitric oxide bioavailability. Genetic deletion of the adaptor protein p66(Shc) protects mice against age-related and hyperglycemia-induced endothelial dysfunction, as well as atherosclerosis and stroke. Furthermore, p66(Shc) mediates vascular dysfunction in hypertensive mice. However, the direct role of p66(Shc) in mediating mechanical force-induced free radical production is unknown; thus, we studied the effect of cyclic stretch on p66(Shc) activation in primary human aortic endothelial cells and aortic endothelial cells isolated from normotensive and hypertensive rats. Exposure of human aortic endothelial cells to cyclic stretch led to a stretch- and time-dependent p66(Shc) phosphorylation at Ser36 downstream of integrin α5ß1 and c-Jun N-terminal kinase. In parallel, nicotinamide adenine dinucleotide phosphate oxidase activation, as well as production of reactive oxygen species, increased, whereas nitric oxide bioavailability decreased. Silencing of p66(Shc) blunted stretch-increased superoxide anion production and nicotinamide adenine dinucleotide phosphate oxidase activation and restored nitric oxide bioavailability. In line with the above, activation of p66(Shc) increased in isolated aortic endothelial cells of spontaneously hypertensive rats compared with normotensive ones. Pathological stretch by activating integrin α5ß1 and c-Jun N-terminal kinase phosphorylates p66(Shc) at Ser36, augments reactive oxygen species production via nicotinamide adenine dinucleotide phosphate oxidase, and in turn reduces nitric oxide bioavailability. This novel molecular pathway may be relevant for endothelial dysfunction and vascular disease in hypertension.


Assuntos
Aorta/metabolismo , Endotélio Vascular/metabolismo , Hipertensão/metabolismo , Proteínas Adaptadoras da Sinalização Shc/metabolismo , Animais , Aorta/fisiopatologia , Células Endoteliais/metabolismo , Endotélio Vascular/fisiopatologia , Humanos , Hipertensão/genética , Hipertensão/fisiopatologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Estresse Oxidativo , Fosforilação , Ratos , Ratos Endogâmicos SHR , Espécies Reativas de Oxigênio/metabolismo , Proteínas Adaptadoras da Sinalização Shc/genética
7.
PLoS One ; 8(1): e53106, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23382833

RESUMO

BACKGROUND: Inflammation plays a key role in atherosclerosis. Sirt1 regulates transcription factors involved in inflammatory processes and blunts atherosclerosis in mice. However, its role in humans remains to be defined. This study was therefore designed to investigate the role of Sirt1 in the development of atherosclerosis. METHODS AND RESULTS: 48 male subjects admitted for cardiac catheterization were subdivided into healthy subjects, patients with stable coronary artery disease (CAD), and with acute coronary syndromes (ACS). Monocytes were isolated and Sirt1 mRNA levels were determined. Sirt1 gene expression was higher in healthy subjects as compared to patients with CAD or ACS (P<0.05), respectively. Interestingly, HDL levels correlated positively with Sirt1 expression. Thus, HDL from the three groups was isolated and incubated with THP-1 monocytes to determine the effects of HDL on Sirt1 protein in controlled experimental conditions. HDL from healthy subjects stimulated Sirt1 expression in THP-1 monocytes to a higher degree than HDL from CAD and ACS patients (P<0.05). Paraoxonase-1 (PON-1), a HDL-associated enzyme, showed a reduced activity in HDL isolated from CAD and ACS patients as compared to the controls (P<0.001). CONCLUSIONS: Monocytic Sirt1 expression is reduced in patients with stable CAD and ACS. Experiments on THP-1 monocytes suggest that this effect is HDL-dependent and is mediated by a reduced activity of HDL-associated enzyme PON1.


Assuntos
Doença da Artéria Coronariana/sangue , Lipoproteínas HDL/sangue , Monócitos/metabolismo , Sirtuína 1/sangue , Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/fisiopatologia , Idoso , Arildialquilfosfatase/genética , Arildialquilfosfatase/metabolismo , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/fisiopatologia , Regulação da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Sirtuína 1/genética
8.
Eur Heart J ; 34(2): 96-103, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23008506

RESUMO

AIMS: Stroke is a leading cause of morbidity and mortality, and its incidence increases with age. Both in animals and in humans, oxidative stress appears to play an important role in ischaemic stroke, with or without reperfusion. The adaptor protein p66(Shc) is a key regulator of reactive oxygen species (ROS) production and a mediator of ischaemia/reperfusion damage in ex vivo hearts. Hence, we hypothesized that p66(Shc) may be involved in ischaemia/reperfusion brain damage. To this end, we investigated whether genetic deletion of p66(Shc) protects from ischaemia/reperfusion brain injury. METHODS AND RESULTS: Transient middle cerebral artery occlusion (MCAO) was performed to induce ischaemia/reperfusion brain injury in wild-type (Wt) and p66(Shc) knockout mice (p66(Shc-/-)), followed by 24 h of reperfusion. Cerebral blood flow and blood pressure measurements revealed comparable haemodynamics in both experimental groups. Neuronal nuclear antigen immunohistochemical staining showed a significantly reduced stroke size in p66(Shc-/-) when compared with Wt mice (P < 0.05, n = 7-8). In line with this, p66(Shc-/-) mice exhibited a less impaired neurological function and a decreased production of free radicals locally and systemically (P < 0.05, n = 4-5). Following MCAO, protein levels of gp91phox nicotinamide adenine dinucleotide phosphate oxidase subunit were increased in brain homogenates of Wt (P < 0.05, n = 4), but not of p66(Shc-/-) mice. Further, reperfusion injury in Wt mice induced p66(Shc) protein in the basilar and middle cerebral artery, but not in brain tissue, suggesting a predominant involvement of vascular p66(Shc). CONCLUSION: In the present study, we show that the deletion of the ageing gene p66(Shc) protects mice from ischaemia/reperfusion brain injury through a blunted production of free radicals. The ROS mediator p66(Shc) may represent a novel therapeutical target for the treatment of ischaemic stroke.


Assuntos
Deleção de Genes , Traumatismo por Reperfusão/genética , Proteínas Adaptadoras da Sinalização Shc/genética , Acidente Vascular Cerebral/genética , Animais , Velocidade do Fluxo Sanguíneo/fisiologia , Pressão Sanguínea/fisiologia , Encefalopatias/fisiopatologia , Circulação Cerebrovascular/fisiologia , Infarto da Artéria Cerebral Média/genética , Infarto da Artéria Cerebral Média/fisiopatologia , Masculino , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , NADPH Oxidase 2 , NADPH Oxidases/metabolismo , Estresse Oxidativo/genética , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Acidente Vascular Cerebral/fisiopatologia
9.
Atherosclerosis ; 220(1): 282-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22100252

RESUMO

OBJECTIVE: The interplay between oxidative stress and inflammation is crucial in the pathogenesis of atherosclerosis. The adaptor protein p66Shc is implicated in atherogenesis and oxidative stress related responses in animal models of diseases. However, its role in humans remains to be defined. In this study, we hypothesized that expression of p66Shc increases in peripheral blood monocytes of patients affected by acute coronary syndromes (ACS). METHODS: Male subjects aged 59±4 (mean±SD) years admitted for cardiac catheterization were subdivided in three groups: (a) no local stenosis for the control group, (b) at least one stenosis ≥75% in either left, circumflex or right coronary artery for the coronary artery disease (CAD) group or (c) ST-elevation/non-ST-elevation myocardial infarction for the ACS group. Monocytes were isolated from whole blood and p66Shc RNA levels were determined by quantitative real time PCR. RESULTS: p66Shc RNA levels were increased in ACS patients as compared to CAD (p=0.007) and controls (p=0.0249). Furthermore, malondialdehyde (MDA) and C-reactive protein (CRP) were increased in plasma of ACS patients. Levels of MDA correlated positively to p66Shc (r=0.376, p=0.01). Our data demonstrate increased p66Shc levels in monocytes of ACS but not CAD patients. CONCLUSION: This study suggests an involvement of p66Shc in the transition of a stable CAD to an ACS patient. p66Shc was associated with states of increased oxidative stress. Further work is needed to understand whether p66Shc may represent a possible pharmacological target or whether it represents an interesting novel biomarker.


Assuntos
Síndrome Coronariana Aguda/genética , Doença da Artéria Coronariana/genética , Monócitos/química , Proteínas Adaptadoras da Sinalização Shc/genética , Síndrome Coronariana Aguda/sangue , Síndrome Coronariana Aguda/diagnóstico por imagem , Distribuição de Qui-Quadrado , Angiografia Coronária , Doença da Artéria Coronariana/sangue , Doença da Artéria Coronariana/diagnóstico por imagem , Humanos , Peroxidação de Lipídeos/genética , Masculino , Malondialdeído/sangue , Pessoa de Meia-Idade , Estresse Oxidativo/genética , RNA Mensageiro/sangue , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Suíça , Regulação para Cima
10.
Cardiovasc Res ; 89(2): 464-72, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-20978007

RESUMO

AIMS: The mammalian silent information regulator-two 1 (Sirt1) blunts the noxious effects of cardiovascular risk factors such as type 2 diabetes mellitus and obesity. Nevertheless, the role of Sirt1 in regulating the expression of tissue factor (TF), the key trigger of coagulation, and arterial thrombus formation remains unknown. METHODS AND RESULTS: Human as well as mouse cell lines were used for in vitro experiments, and C57Bl/6 mice for in vivo procedures. Sirt1 inhibition by splitomicin or sirtinol enhanced cytokine-induced endothelial TF protein expression as well as surface activity, while TF pathway inhibitor protein expression did not change. Sirt1 inhibition further enhanced TF mRNA expression, TF promoter activity, and nuclear translocation as well as DNA binding of the p65 subunit of nuclear factor-kappa B (NFκB/p65). Sirt1 siRNA enhanced TF protein and mRNA expression, and this effect was reduced in NFκB/p65(-/-) mouse embryonic fibroblasts reconstituted with non-acetylatable Lys(310)-mutant NFκB/p65. Activation of the mitogen-activated protein kinases p38, c-Jun NH(2)-terminal kinase, and p44/42 (ERK) remained unaffected. In vivo, mice treated with the Sirt1 inhibitor splitomicin exhibited enhanced TF activity in the arterial vessel wall and accelerated carotid artery thrombus formation in a photochemical injury model. CONCLUSION: We provide pharmacological and genetic evidence that Sirt1 inhibition enhances TF expression and activity by increasing NFκB/p65 activation in human endothelial cells. Furthermore, Sirt1 inhibition induces arterial thrombus formation in vivo. Hence, modulation of Sirt1 may offer novel therapeutic options for targeting thrombosis.


Assuntos
Células Endoteliais/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Sirtuína 1/antagonistas & inibidores , Sirtuína 1/metabolismo , Tromboplastina/metabolismo , Trombose/etiologia , Animais , Benzamidas/farmacologia , Sítios de Ligação , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Células Endoteliais/enzimologia , Ativadores de Enzimas , Genes Reporter , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Naftalenos/farmacologia , Naftóis/farmacologia , Regiões Promotoras Genéticas , Pironas/farmacologia , Interferência de RNA , RNA Mensageiro/metabolismo , Resveratrol , Sirtuína 1/deficiência , Sirtuína 1/genética , Estilbenos/farmacologia , Tromboplastina/genética , Trombose/sangue , Trombose/enzimologia , Trombose/genética , Fator de Transcrição RelA/deficiência , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/metabolismo , Transfecção
11.
Arterioscler Thromb Vasc Biol ; 30(10): 1960-7, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20689076

RESUMO

OBJECTIVE: To assess the effects of aging on arterial thrombus formation by comparing 2-year-old with 11-week-old C57Bl6 mice. METHODS AND RESULTS: Aging is a major risk factor for cardiovascular disease. In humans, assessing the direct effects of aging on vascular homeostasis is difficult because it occurs in the presence of other risk factors. Arterial thrombosis is the critical event in cardiovascular diseases; however, it is not known whether aging per se promotes its occurrence. Mice represent an interesting system to address this issue because they age without spontaneously developing other risk factors. Organ chamber experiments confirmed the advanced level of aging of old mice. As previously shown, old mice exhibited endothelial dysfunction; however, arterial thrombosis induced by photochemical injury was unchanged. Arterial tissue factor expression and activity; expressions of tissue factor pathway inhibitor, thrombomodulin, and plasminogen activator inhibitor 1; prothrombin time; partial thromboplastin time; thrombin-antithrombin complex; and platelet activation were comparable in both groups. CONCLUSIONS: Although these results cannot be directly extrapolated to humans, this study contributes novel important information on the direct effect of aging on arterial thrombosis and underscores the importance of controlling modifiable risk factors in aged individuals.


Assuntos
Envelhecimento/patologia , Envelhecimento/fisiologia , Trombose das Artérias Carótidas/fisiopatologia , Endotélio Vascular/fisiopatologia , Envelhecimento/genética , Animais , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Doenças da Aorta/etiologia , Doenças da Aorta/patologia , Doenças da Aorta/fisiopatologia , Sequência de Bases , Coagulação Sanguínea , Plaquetas/fisiologia , Trombose das Artérias Carótidas/etiologia , Trombose das Artérias Carótidas/patologia , Primers do DNA/genética , Modelos Animais de Doenças , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fatores de Risco , Tromboplastina/genética , Tromboplastina/metabolismo , Vasodilatação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...