Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Pharmacol Exp Ther ; 325(3): 782-90, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18354059

RESUMO

Mitochondrial reactive oxygen species (ROS) and endothelial dysfunction are key contributors to cerebrovascular pathophysiology. We previously found that 17beta-estradiol profoundly affects mitochondrial function in cerebral blood vessels, enhancing efficiency of energy production and suppressing mitochondrial oxidative stress. To determine whether estrogen specifically affects endothelial mitochondria through receptor mechanisms, we used cultured human brain microvascular endothelial cells (HBMECs). 17beta-Estradiol treatment for 24 h increased mitochondrial cytochrome c protein and mRNA; use of silencing RNA for estrogen receptors (ERs) showed that this effect involved ERalpha, but not ERbeta. Mitochondrial ROS were determined by measuring the activity of aconitase, an enzyme with an iron-sulfur center inactivated by mitochondrial superoxide. 17beta-Estradiol increased mitochondrial aconitase activity in HBMECs, indicating a reduction in ROS. Direct measurement of mitochondrial superoxide with MitoSOX Red showed that 17beta-estradiol, but not 17alpha-estradiol, significantly decreased mitochondrial superoxide production, an effect blocked by the ER antagonist, ICI-182,780 (fulvestrant). Selective ER agonists demonstrated that the decrease in mitochondrial superoxide was mediated by ERalpha, not ERbeta. The selective estrogen receptor modulators, raloxifene and 4-hydroxy-tamoxifen, differentially affected mitochondrial superoxide production, with raloxifene acting as an agonist but 4-hydroxy-tamoxifen acting as an estrogen antagonist. Changes in superoxide by 17beta-estradiol could not be explained by changes in manganese superoxide dismutase. Instead, ERalpha-mediated decreases in mitochondrial ROS may depend on the concomitant increase in mitochondrial cytochrome c, previously shown to act as an antioxidant. Mitochondrial protective effects of estrogen in cerebral endothelium may contribute to sex differences in the occurrence of stroke and other age-related neurodegenerative diseases.


Assuntos
Células Endoteliais/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio/metabolismo , Estrogênios/farmacologia , Mitocôndrias/efeitos dos fármacos , Aconitato Hidratase/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Linhagem Celular , Citocromos c/genética , Células Endoteliais/metabolismo , Receptor alfa de Estrogênio/genética , Fumarato Hidratase/metabolismo , Humanos , Mitocôndrias/metabolismo , Interferência de RNA , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Superóxido Dismutase/metabolismo
2.
Am J Physiol Heart Circ Physiol ; 292(5): H2333-40, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17208996

RESUMO

In young adult females, estrogen treatment suppresses the cerebrovascular inflammatory response; this is mediated in part via NF-kappaB, a key regulator of inflammatory genes. To examine whether age modifies effects of estrogen on vascular inflammation in the brain, female rats, 3 and 12 mo of age, were ovariectomized; half were treated with estrogen for 4 wk. Cerebral blood vessels were isolated from the animals at 4 and 13 mo of age. Inflammation was induced by LPS, either injected in vivo or incubated with isolated vessels ex vivo. Basal levels of cytoplasmic NF-kappaB were significantly higher in cerebral vessels of young rats, but the ratio of nuclear to cytoplasmic levels was greater in middle-aged animals. LPS exposure increased nuclear NF-kappaB DNA binding activity, protein levels of inducible nitric oxide synthase and cyclooxygenase-2, and production of nitric oxide and PGE(2) in cerebral vessels. All effects of LPS were markedly greater in vessels from the older animals. Estrogen significantly inhibited the LPS-induced increase in NF-kappaB DNA binding activity in cerebral vessels from animals at both ages. In 4-mo-old rats, estrogen also significantly suppressed LPS induction of inducible nitric oxide synthase and cyclooxygenase-2 proteins, as well as production of nitric oxide and PGE(2). In contrast, in 13-mo-old females, estrogen did not significantly affect these indexes of cerebrovascular inflammation. Thus the protective, anti-inflammatory effect of estrogen on cerebral blood vessels that is observed in young adults may be attenuated in aged animals, which exhibit a greater overall cerebrovascular response to inflammatory stimuli.


Assuntos
Envelhecimento/metabolismo , Citocinas/metabolismo , Estradiol/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Vasculite do Sistema Nervoso Central/tratamento farmacológico , Vasculite do Sistema Nervoso Central/metabolismo , Envelhecimento/efeitos dos fármacos , Animais , Feminino , Terapia de Reposição Hormonal/métodos , Ovariectomia , Ratos , Ratos Endogâmicos F344 , Resultado do Tratamento
3.
Am J Physiol Endocrinol Metab ; 291(2): E261-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16492687

RESUMO

The potential benefit of ovarian hormone replacement therapy in cerebrovascular disease is well supported by experimental observations but not by recent large, randomized clinical trials. This discrepancy points out the need for better understanding of the vascular actions of ovarian hormones as well as medroxyprogesterone acetate (MPA), a synthetic analog of progesterone (P) widely prescribed in combination with estrogens. Therefore, we investigated whether in vivo exposure to 17beta-estradiol (E) and/or P or MPA modifies inflammation in the cerebral vasculature, a key process in the evolution of ischemic brain injury. Female rats were injected (ip) with LPS to induce inflammation, and 6 h later brains were taken for blood vessel isolation and Western blot analysis of the inflammatory enzymes inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2). In ovariectomized (O) females, LPS induced cerebrovascular iNOS and COX-2; however, this effect was significantly decreased when O animals were treated for 3 wk with E. In contrast, treatment of O females with either MPA or P exacerbated the cerebrovascular inflammatory response to LPS. In intact females, LPS induction of iNOS and COX-2 in cerebral vessels was found to vary with the stage of the estrous cycle: LPS had the greatest effect during estrus, when circulating estrogen is low and progesterone is high. Thus exposure to endogenous or exogenous ovarian hormones appears to modulate cerebrovascular inflammation. Anti-inflammatory effects of estrogen would attenuate ischemic brain injury; however, this vasoprotective benefit may be diminished in the presence of progestagens.


Assuntos
Citocinas/metabolismo , Estradiol/administração & dosagem , Progesterona/administração & dosagem , Vasculite do Sistema Nervoso Central/metabolismo , Animais , Relação Dose-Resposta a Droga , Feminino , Fatores Imunológicos , Lipopolissacarídeos , Acetato de Medroxiprogesterona/administração & dosagem , Ratos , Ratos Endogâmicos F344 , Vasculite do Sistema Nervoso Central/induzido quimicamente
4.
Eur J Pharmacol ; 478(1): 35-8, 2003 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-14555182

RESUMO

The enzyme endothelial nitric oxide synthase (eNOS) plays a critical role in the maintenance of vascular tone. The mechanism by which estrogen increases eNOS function remains controversial. We demonstrate here using real-time polymerase chain reaction (PCR) and immunoblot analysis that in vivo estrogen treatment leads to a 100% increase in eNOS messenger RNA (mRNA) copy number and increases eNOS protein levels by 47% in mouse cerebral blood vessels. These data suggest that estrogen can modulate eNOS at the transcriptional level in blood vessels in vivo.


Assuntos
Circulação Cerebrovascular/efeitos dos fármacos , Estradiol/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Óxido Nítrico Sintase/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Animais , Encéfalo/irrigação sanguínea , Encéfalo/efeitos dos fármacos , Encéfalo/enzimologia , Circulação Cerebrovascular/fisiologia , Feminino , Regulação Enzimológica da Expressão Gênica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Ovariectomia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...