Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Med ; 7(7): 853-7, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11433352

RESUMO

Atherosclerosis is the primary cause of cardiovascular disease, and the risk for atherosclerosis is inversely proportional to circulating levels of high-density lipoprotein (HDL) cholesterol. However, the mechanisms by which HDL is atheroprotective are complex and not well understood. Here we show that HDL stimulates endothelial nitric oxide synthase (eNOS) in cultured endothelial cells. In contrast, eNOS is not activated by purified forms of the major HDL apolipoproteins ApoA-I and ApoA-II or by low-density lipoprotein. Heterologous expression experiments in Chinese hamster ovary cells reveal that scavenger receptor-BI (SR-BI) mediates the effects of HDL on the enzyme. HDL activation of eNOS is demonstrable in isolated endothelial-cell caveolae where SR-BI and eNOS are colocalized, and the response in isolated plasma membranes is blocked by antibodies to ApoA-I and SR-BI, but not by antibody to ApoA-II. HDL also enhances endothelium- and nitric-oxide-dependent relaxation in aortae from wild-type mice, but not in aortae from homozygous null SR-BI knockout mice. Thus, HDL activates eNOS via SR-BI through a process that requires ApoA-I binding. The resulting increase in nitric-oxide production might be critical to the atheroprotective properties of HDL and ApoA-I.


Assuntos
Antígenos CD36/metabolismo , Lipoproteínas HDL/metabolismo , Proteínas de Membrana , Óxido Nítrico Sintase/metabolismo , Receptores Imunológicos , Receptores de Lipoproteínas , Animais , Sequência de Bases , Antígenos CD36/genética , Antígenos CD36/fisiologia , Células CHO , Linhagem Celular Transformada , Cricetinae , Primers do DNA , Ativação Enzimática , Óxido Nítrico Sintase Tipo III , Ligação Proteica , Receptores Depuradores , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Receptores Depuradores Classe B , Ovinos
2.
Nat Med ; 3(5): 545-8, 1997 May.
Artigo em Inglês | MEDLINE | ID: mdl-9142124

RESUMO

The atheroprotective effects of estrogen in women are well recognized, but the underlying mechanisms responsible are not well understood. Blood vessel cells express the classic estrogen receptor, ER alpha (ref. 2-6), and are directly affected by estrogen, which inhibits the development of atherosclerotic and injury-induced vascular lesions. We have generated mice in which the ER alpha gene is disrupted and have used a mouse model of carotid arterial injury to compare the effects of estrogen on wild-type and estrogen receptor-deficient mice. Increases in vascular medial area and smooth muscle cell proliferation were quantified following vascular injury in ovariectomized mice treated with vehicle or with physiologic levels of 17 beta-estradiol. Surprisingly, in both wild-type and estrogen receptor-deficient mice, 17 beta-estradiol markedly inhibited to the same degree all measures of vascular injury. These data demonstrate that estrogen inhibits vascular by a novel mechanism that is independent of the classic estrogen receptor, ER alpha.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Estradiol/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Receptores de Estrogênio/fisiologia , Animais , Artérias Carótidas , Divisão Celular , Endotélio Vascular/patologia , Feminino , Expressão Gênica , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/patologia , Receptores de Estrogênio/genética
3.
Science ; 286(5444): 1583-7, 1999 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-10567269

RESUMO

Contraction and relaxation of smooth muscle are regulated by myosin light-chain kinase and myosin phosphatase through phosphorylation and dephosphorylation of myosin light chains. Cyclic guanosine monophosphate (cGMP)-dependent protein kinase Ialpha (cGKIalpha) mediates physiologic relaxation of vascular smooth muscle in response to nitric oxide and cGMP. It is shown here that cGKIalpha is targeted to the smooth muscle cell contractile apparatus by a leucine zipper interaction with the myosin-binding subunit (MBS) of myosin phosphatase. Uncoupling of the cGKIalpha-MBS interaction prevents cGMP-dependent dephosphorylation of myosin light chain, demonstrating that this interaction is essential to the regulation of vascular smooth muscle cell tone.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Músculo Liso Vascular/enzimologia , Cadeias Leves de Miosina/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Células Cultivadas , Proteína Quinase Dependente de GMP Cíclico Tipo I , Proteínas Quinases Dependentes de GMP Cíclico/química , Proteínas Quinases Dependentes de GMP Cíclico/genética , Histonas/metabolismo , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Zíper de Leucina , Contração Muscular , Relaxamento Muscular , Músculo Liso Vascular/fisiologia , Mutagênese Sítio-Dirigida , Fosfatase de Miosina-de-Cadeia-Leve , Fosfoproteínas Fosfatases/química , Fosforilação , Testes de Precipitina , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Especificidade por Substrato , Transfecção , Técnicas do Sistema de Duplo-Híbrido
4.
Climacteric ; 12 Suppl 1: 18-21, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19811235

RESUMO

This brief review summarizes the current state of the field for estrogen receptor actions in the cardiovascular system and the cardiovascular effects of hormone replacement therapy (HRT). It is organized into three parts: a short Introduction and overview of the current view of how estrogen works on blood vessels; a summary of the current status of clinical information regarding HRT and cardiovascular effects; and an update on state-of-the-art mouse models of estrogen action using estrogen receptor knockout mice.


Assuntos
Sistema Cardiovascular/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Terapia de Reposição de Estrogênios , Estrogênios/farmacologia , Receptores de Estrogênio/fisiologia , Animais , Fenômenos Fisiológicos Cardiovasculares/efeitos dos fármacos , Sistema Cardiovascular/metabolismo , Endotélio Vascular/metabolismo , Humanos , Camundongos , Camundongos Knockout , Óxido Nítrico/metabolismo , Receptores de Estrogênio/metabolismo
5.
J Clin Invest ; 81(1): 62-8, 1988 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-3335643

RESUMO

The conversion of tissue macrophages into cholesteryl ester-rich foam cells is a crucial early event in atherogenesis. We studied the platelet as a potential source of cholesterol for esterification by macrophages because (a) platelets are rich in free cholesterol, (b) they adhere to macrophages early in atherogenesis, and (c) vascular injury can induce foam cell formation in the absence of hyperlipoproteinemia. We found that washed, activated human platelets from normocholesterolemic donors stimulated cholesteryl ester formation by the human monocyte-derived cell, U-937. Platelet cholesterol, released from platelets activated with calf skin collagen, was approximately equipotent at donating cholesterol to U-937 cells for esterification as normal human low density lipoprotein cholesterol. The stimulation of cholesteryl ester formation by activated human platelets demonstrated both concentration and time dependence. When hypercholesterolemic donors were studied, it was found that increasing plasma levels of cholesterol correlated directly with the ability of these hypercholesterolemic platelets to support cholesteryl ester synthesis by U-937 cells. Cholesterol-donating activity was also found in a 1,000-g supernatants of activated platelets. These observations point to a new and potentially important role for platelets in atherogenesis and suggest a mechanism for foam cell formation in the absence of marked hypercholesterolemia.


Assuntos
Plaquetas/fisiologia , Ésteres do Colesterol/biossíntese , Monócitos/metabolismo , Plaquetas/metabolismo , Linhagem Celular , Colesterol/sangue , Colágeno , Humanos , Lipoproteínas/farmacologia , Monócitos/efeitos dos fármacos , Agregação Plaquetária
6.
J Clin Invest ; 99(4): 570-6, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9045856

RESUMO

A new mouse cardiac electrophysiology method was used to study mice harboring an alpha-myosin heavy chain Arg403Gln missense mutation (alpha-MHC403/+), which results in histological and hemodynamic abnormalities characteristic of familial hypertrophic cardiomyopathy (FHC) and sudden death of uncertain etiology during exercise. Wild-type animals had completely normal cardiac electrophysiology. In contrast, FHC mice demonstrated (a) electrocardiographic abnormalities including prolonged repolarization intervals and rightward axis; (b) electrophysiological abnormalities including heterogeneous ventricular conduction properties and prolonged sinus node recovery time; and (c) inducible ventricular ectopy. These data identify distinct electrophysiologic abnormalities in FHC mice with a specific alpha-myosin mutation, and also validate a novel method to explore in vivo the relationship between specific genotypes and their electrophysiologic phenotypes.


Assuntos
Arritmias Cardíacas/genética , Arritmias Cardíacas/fisiopatologia , Cardiomiopatia Hipertrófica/genética , Cardiomiopatia Hipertrófica/fisiopatologia , Eletrofisiologia , Cadeias Pesadas de Miosina/genética , Animais , Arritmias Cardíacas/etiologia , Estimulação Elétrica , Eletrodos Implantados , Eletrofisiologia/métodos , Sistema de Condução Cardíaco/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Estudos Prospectivos
7.
J Clin Invest ; 101(12): 2851-61, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9637719

RESUMO

The classical estrogen receptor ERalpha mediates many of the known cardiovascular effects of estrogen and is expressed in male and female vascular cells. Estrogen-independent activation of ERalpha is known to occur in cells from reproductive tissues, but has not been investigated previously in vascular cells. In this study, transient transfection assays in human saphenous vein smooth muscle cells (HSVSMC) and pulmonary vein endothelial cells (PVEC) demonstrated ERalpha-dependent activation of estrogen response element-based, and vascular endothelial growth factor-based reporter plasmids by both estrogen-deficient FBS (ED-FBS) and EGF. In nonvascular cells, ERalpha-mediated gene expression can be activated via mitogen-activated protein (MAP) kinase- induced phosphorylation of serine 118 of ERalpha. However, in vascular cells, we found that pharmacologic inhibition of MAP kinase did not alter EGF-mediated ERalpha activation. In addition, a mutant ER containing an alanine-for-serine substitution at position 118 was activated to the same degree as the wild-type receptor by ED-FBS and EGF in both HSVSMC and PVEC. Furthermore, constitutively active MAP kinase kinase (MAPKK) activated ERalpha in Cos1 cells as expected, but MAPKK inhibited ER activation in PVEC. We conclude that growth factors also stimulate ERalpha-mediated gene expression in vascular cells, but find that this occurs via a MAP kinase-independent pathway distinct from that reported previously in nonvascular cells.


Assuntos
Substâncias de Crescimento/farmacologia , Músculo Liso Vascular/fisiologia , Receptores de Estrogênio/fisiologia , Transdução de Sinais/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Células Cultivadas , Endotélio Vascular/fisiologia , Feminino , Humanos , Masculino , Quinases de Proteína Quinase Ativadas por Mitógeno , Proteínas Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos
8.
J Clin Invest ; 103(3): 401-6, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-9927501

RESUMO

Estrogen is an important vasoprotective molecule that causes the rapid dilation of blood vessels by activating endothelial nitric oxide synthase (eNOS) through an unknown mechanism. In studies of intact ovine endothelial cells, 17beta-estradiol (E2) caused acute (five-minute) activation of eNOS that was unaffected by actinomycin D but was fully inhibited by concomitant acute treatment with specific estrogen receptor (ER) antagonists. Overexpression of the known transcription factor ERalpha led to marked enhancement of the acute response to E2, and this was blocked by ER antagonists, was specific to E2, and required the ERalpha hormone-binding domain. In addition, the acute response of eNOS to E2 was reconstituted in COS-7 cells cotransfected with wild-type ERalpha and eNOS, but not by transfection with eNOS alone. Furthermore, the inhibition of tyrosine kinases or mitogen-activated protein (MAP) kinase kinase prevented the activation of eNOS by E2, and E2 caused rapid ER-dependent activation of MAP kinase. These findings demonstrate that the short-term effects of estrogen central to cardiovascular physiology are mediated by ERalpha functioning in a novel, nongenomic manner to activate eNOS via MAP kinase-dependent mechanisms.


Assuntos
Endotélio Vascular/metabolismo , Estrogênios/farmacologia , Óxido Nítrico Sintase/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Ativação Enzimática , Receptor alfa de Estrogênio , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase Tipo III , Receptores de Estrogênio/agonistas , Ovinos , Transdução de Sinais/efeitos dos fármacos
9.
J Clin Invest ; 86(1): 228-34, 1990 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-2195060

RESUMO

The effect of hypercholesterolemia on vascular function was studied in humans. To eliminate the potential confounding effects of atherosclerosis, vascular reactivity was measured in the forearm resistance vessels of 11 normal subjects (serum LDL cholesterol = 111 +/- 7 mg/dl) and 13 patients with hypercholesterolemia (serum LDL cholesterol = 211 +/- 19 mg/dl, P less than 0.05). Each subject received intrabrachial artery infusions of methacholine, which releases endothelium-derived relaxant factor, and nitroprusside which directly stimulates guanylate cyclase in vascular smooth muscle. Maximal vasodilatory potential was determined during reactive hyperemia. Vasoconstrictive responsiveness was examined during intra-arterial phenylephrine infusion. Forearm blood flow was determined by venous occlusion plethysmography. Basal forearm blood flow in normal and hypercholesterolemic subjects was comparable. Similarly, reactive hyperemic blood flow did not differ between the two groups. In contrast, the maximal forearm blood flow response to methacholine in hypercholesterolemic subjects was less than that observed in normal subjects. In addition, the forearm blood flow response to nitroprusside was less in hypercholesterolemic subjects. There was no difference in the forearm vasoconstrictive response to phenylephrine in the two groups. Thus, the vasodilator responses to methacholine and nitroprusside were blunted in patients with hypercholesterolemia. We conclude that in humans with hypercholesterolemia, there is a decreased effect of nitrovasodilators, including endothelium-derived relaxing factor, on the vascular smooth muscle of resistance vessels.


Assuntos
Hipercolesterolemia/fisiopatologia , Vasodilatação/efeitos dos fármacos , Adulto , Braço/irrigação sanguínea , Aspirina/farmacologia , Feminino , Humanos , Masculino , Cloreto de Metacolina , Compostos de Metacolina/farmacologia , Nitroprussiato/farmacologia , Fenilefrina/farmacologia , Fluxo Sanguíneo Regional/efeitos dos fármacos
10.
J Clin Invest ; 108(4): 611-8, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11518735

RESUMO

Clinical studies of hormone replacement therapy to prevent cardiovascular diseases have heightened interest in the cardiovascular effects of progestins. However, the role of the progesterone receptor (PR) in vascular biology has not been studied in vivo. We studied ovariectomized female PR knockout (PRKO) mice and their wild-type (WT) littermates using the mouse carotid artery injury model. Placebo-treated PRKO mice showed significantly greater vascular medial hypertrophy and vascular smooth muscle cell (VSMC) proliferation in response to vascular injury than did WT mice. Progesterone had no significant effect in the PRKO mice, but worsened the response to injury in WT mice. VSMCs cultured from PRKO mouse aortae were markedly hyperproliferative, and their growth was not affected by progesterone. In contrast to the in vivo findings, progesterone inhibited proliferation of WT-derived VSMCs. Furthermore, reintroduction of PR into PRKO-derived VSMCs using adenoviral methods restored progesterone-mediated inhibition of proliferation to these cells. This effect was reversed by the PR antagonist, RU 486. Thus, the effects of PR and progesterone differ markedly between cultured VSMCs and intact blood vessels. These data demonstrate a direct role for the PR in regulating the response to vascular injury and VSMC proliferation.


Assuntos
Lesões das Artérias Carótidas , Receptores de Progesterona/fisiologia , Animais , Artéria Carótida Primitiva/patologia , Divisão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Replicação do DNA/efeitos dos fármacos , Feminino , Antagonistas de Hormônios/farmacologia , Hiperplasia , Camundongos , Camundongos Knockout , Mifepristona/farmacologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/patologia , Ovariectomia , Progesterona/antagonistas & inibidores , Progesterona/farmacologia , Receptores de Progesterona/deficiência , Receptores de Progesterona/efeitos dos fármacos , Receptores de Progesterona/genética , Proteínas Recombinantes de Fusão/fisiologia , Transfecção
11.
J Clin Invest ; 96(5): 2482-8, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-7593638

RESUMO

The atheroprotective effects of estrogen are well documented, but the mechanisms responsible for these effects are not well understood. To study the role of physiologic (nanomolar) estrogen levels on the arterial response-to-injury, we applied a mouse carotid artery injury model to ovariectomized C57BL/6J mice. Mice were treated with vehicle (-E2, n = 10) or 17 beta-estradiol (+E2, n = 10) for 7 d, subjected to unilateral carotid injury, and 14 d later contralateral (normal = NL) and injured carotids from -E2 and +E2 animals were pressure fixed, harvested, and analyzed by quantitative morphometry. E2 levels in +E2 mice were consistently in the nanomolar range (2.1-2.5 nM) at days 0, 7, and 14. At 14 d, measures of both intimal and medial area were markedly increased in the -E2 group: (-E2 vs NL, P < 0.05 for both), but were unchanged from normal levels in the +E2 group (+E2 vs NL, P = NS and +E2 vs -E2, P < 0.05 for both). Cellular proliferation, as assessed by bromodeoxyuridine (BrdU) labeling, was significantly increased over NL in the -E2 mice, but this increase was markedly attenuated in the estrogen replacement group (total BrdU positive cells/section: NL = 6.4 +/- 4.5; -E2 = 113 +/- 26, +E2 = 40 +/- 3.7; -E2 vs NL, P < 0.05; +E2 vs NL, P = NS; -E2 vs +E2, P < 0.05). These data (a) demonstrate significant suppression of the mouse carotid response-to-injury by physiologic levels of estrogen replacement; (b) support the utility of this model in the study of the biologic effects of estrogen on the vascular-injury response; and (c) suggest a direct effect of estrogen on vascular smooth muscle cell proliferation in injured vessels.


Assuntos
Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Estradiol/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Feminino , Hiperplasia/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Túnica Íntima/patologia , Túnica Média/patologia
12.
J Clin Invest ; 103(4): R1-7, 1999 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10021468

RESUMO

Myotonic dystrophy (DM) is the most common form of muscular dystrophy and is caused by expansion of a CTG trinucleotide repeat on human chromosome 19. Patients with DM develop atrioventricular conduction disturbances, the principal cardiac manifestation of this disease. The etiology of the pathophysiological changes observed in DM has yet to be resolved. Haploinsufficiency of myotonic dystrophy protein kinase (DMPK), DM locus-associated homeodomain protein (DMAHP) and/or titration of RNA-binding proteins by expanded CUG sequences have been hypothesized to underlie the multi-system defects observed in DM. Using an in vivo murine electrophysiology study, we show that cardiac conduction is exquisitely sensitive to DMPK gene dosage. DMPK-/- mice develop cardiac conduction defects which include first-, second-, and third-degree atrioventricular (A-V) block. Our results demonstrate that the A-V node and the His-Purkinje regions of the conduction system are specifically compromised by DMPK loss. Importantly, DMPK+/- mice develop first-degree heart block, a conduction defect strikingly similar to that observed in DM patients. These results demonstrate that DMPK dosage is a critical element modulating cardiac conduction integrity and conclusively link haploinsufficiency of DMPK with cardiac disease in myotonic dystrophy.


Assuntos
Dosagem de Genes , Bloqueio Cardíaco/genética , Distrofia Miotônica/genética , Proteínas Serina-Treonina Quinases/fisiologia , Animais , Colinérgicos , Modelos Animais de Doenças , Feminino , Bloqueio Cardíaco/fisiopatologia , Masculino , Camundongos , Mutagênese , Miocárdio/patologia , Distrofia Miotônica/fisiopatologia , Miotonina Proteína Quinase , Proteínas Serina-Treonina Quinases/genética , Simpatolíticos
13.
Circ Res ; 87(11): 956-60, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-11090536

RESUMO

The administration of estrogens in animals tends to inhibit the development of atherosclerosis. [Therefore], attempts are being made in human males to prevent further progress of the disease in cases of angina or previous coronary thrombosis by long-range estrogen therapy. This work is in its infancy. It is hoped that, with further research, compounds might be found that eliminate the undesirable action of such hormones and yet retain its beneficial effects on arteries (Samuel A. Levine, 1958).


Assuntos
Retículo Endoplasmático/enzimologia , Óxido Nítrico Sintase/metabolismo , Animais , Doenças Cardiovasculares/enzimologia , Doenças Cardiovasculares/prevenção & controle , Ativação Enzimática/fisiologia , Estrogênios/uso terapêutico , Feminino , Proteínas de Choque Térmico HSP90/metabolismo , Terapia de Reposição Hormonal , Humanos , Masculino , Óxido Nítrico Sintase Tipo III , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Subunidades Proteicas , Receptores de Estrogênio/metabolismo , Transdução de Sinais
14.
Circ Res ; 89(6): 534-9, 2001 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-11557741

RESUMO

The two known estrogen receptors, ERalpha and ERbeta, mediate the effects of estrogen in all target tissues, including blood vessels. We have shown previously that estrogen inhibits vascular injury response to the same extent in female wild-type (WT), ERalpha knockout (ERalphaKO(CH)), and ERbeta knockout (ERbetaKO(CH)) mice. We generated mice harboring disruptions of both ERalpha and ERbeta genes (ERalpha,betaKO(CH)) by breeding and studied the effect of 17beta-estradiol (E2) on vascular injury responses in ovariectomized female ERalpha,betaKO(CH) mice and WT littermates. E2 inhibited increases in vascular medial area following injury in the WT mice but not in the ERalpha,betaKO(CH) mice, demonstrating for the first time that the two known estrogen receptors are necessary and sufficient to mediate estrogen inhibition of a component of the vascular injury response. Surprisingly, as in WT littermates, E2 still significantly increased uterine weight and inhibited vascular smooth muscle cell (VSMC) proliferation following injury in the ERalpha,betaKO(CH) mice. These data support that the role of estrogen receptors differs for specific components of the vascular injury response in the ERalpha,betaKO(CH) mice. The results leave unresolved whether E2 inhibition of VSMC proliferation in ERalpha,betaKO(CH) mice is caused by a receptor-independent mechanism, an unidentified receptor responsive to estrogen, or residual activity of the ERalpha splice variant reported previously in the parental ERalphaKO(CH) mice. These possibilities may be resolved by studies of mice in which ERalpha has been fully disrupted (ERalphaKO(St)), which are in progress.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Estrogênios/uso terapêutico , Receptores de Estrogênio/genética , Animais , Bromodesoxiuridina/metabolismo , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/patologia , Lesões das Artérias Carótidas/genética , Divisão Celular/efeitos dos fármacos , Estradiol/uso terapêutico , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Genótipo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Ovariectomia
15.
Circ Res ; 87(11): E44-52, 2000 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-11090554

RESUMO

Estrogen causes nitric oxide (NO)-dependent vasodilation due to estrogen receptor (ER) alpha-mediated, nongenomic activation of endothelial NO synthase (eNOS). The subcellular site of interaction between ERalpha and eNOS was determined in studies of isolated endothelial cell plasma membranes. Estradiol (E(2), 10(-8) mol/L) caused an increase in eNOS activity in plasma membranes in the absence of added calcium, calmodulin, or eNOS cofactors, which was blocked by ICI 182,780 and ERalpha antibody. Immunoidentification studies detected the same 67-kDa protein in endothelial cell nucleus, cytosol, and plasma membrane. Plasma membranes from COS-7 cells expressing eNOS and ERalpha displayed ER-mediated eNOS stimulation, whereas membranes from cells expressing eNOS alone or ERalpha plus a myristoylation-deficient mutant eNOS were insensitive. Fractionation of endothelial cell plasma membranes revealed ERalpha protein in caveolae, and E(2) caused stimulation of eNOS in isolated caveolae that was ER-dependent; noncaveolae membranes were insensitive. Acetylcholine and bradykinin also activated eNOS in isolated caveolae. Furthermore, the effect of E(2) on eNOS in caveolae was prevented by calcium chelation. Thus, a subpopulation of ERalpha is localized to endothelial cell caveolae where they are coupled to eNOS in a functional signaling module that may regulate the local calcium environment. The full text of this article is available at http://www.circresaha.org.


Assuntos
Cavéolas/metabolismo , Óxido Nítrico Sintase/metabolismo , Receptores de Estrogênio/metabolismo , Transdução de Sinais , Acetilcolina/farmacologia , Animais , Células COS , Cálcio/metabolismo , Calmodulina/metabolismo , Caveolina 1 , Caveolinas/metabolismo , Membrana Celular/enzimologia , Células Cultivadas , Quelantes , Colinérgicos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Estradiol/farmacologia , Receptor alfa de Estrogênio , Immunoblotting , Óxido Nítrico Sintase Tipo III , Ovinos , Transdução de Sinais/efeitos dos fármacos
16.
Biochim Biophys Acta ; 1137(3): 248-56, 1992 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-1332779

RESUMO

The biochemical responses of intact human platelets to the monoclonal antibody (mAb) AG-1 were investigated. AG-1 is a murine IgG mAb that recognizes a series of platelet membrane glycoproteins (Gp) from M(r) 21,000 to 29,000, one of which is the M(r) 24,000 (p24) receptor for anti-CD9 mAbs. AG-1 causes platelet aggregation and secretion. Platelets binding AG-1 demonstrate a dose- and time-dependent breakdown of phosphatidylinositol 4,5-bisphosphate (PIP2), production of diacylglycerol, and generation of phosphatidic acid (PA). These events are associated with the activation of protein kinase C (PKC), an increase in intracellular calcium, and fibrinogen binding. Platelet PA generation and PKC activation in response to AG-1 are inhibited by mAbs to platelet GpIIb-IIIa or by extracellular EGTA, but not by a mAb to platelet GpIb or by inhibiting platelet Na+/H+ exchange with 5-(N-ethyl-N-isopropyl)amiloride. Platelet cytoplasmic free calcium ([Ca2+]i) is elevated in response to AG-1, and this elevation is inhibited by mAbs to GpIIb-IIIa, an RGDS peptide or by chelating extracellular calcium. These results suggest that AG-1 binding to a unique platelet-surface glycoprotein initiates platelet responses through the activation of PIP2-specific phospholipase C, and that this occurs through a signal pathway that is dependent on GpIIb-IIIa and extracellular calcium.


Assuntos
Anticorpos Monoclonais , Cálcio/metabolismo , Glicoproteínas da Membrana de Plaquetas/metabolismo , Sequência de Aminoácidos , Cátions Bivalentes , Ativação Enzimática , Fibrinogênio/metabolismo , Humanos , Dados de Sequência Molecular , Oligopeptídeos/farmacologia , Ácidos Fosfatídicos/biossíntese , Fosfatidilinositol 4,5-Difosfato , Fosfatidilinositóis/biossíntese , Fosforilação , Inibidores da Agregação Plaquetária/farmacologia , Glicoproteínas da Membrana de Plaquetas/imunologia , Proteína Quinase C/metabolismo , Transdução de Sinais
17.
J Am Coll Cardiol ; 37(1): 195-200, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11153738

RESUMO

OBJECTIVES: The goal of this study was to examine the effect of heart failure etiology on peripheral vascular endothelial function in cardiac transplant recipients. BACKGROUND: Peripheral vascular endothelial dysfunction occurs in patients with heart failure of either ischemic or nonischemic etiology. The effect of heart failure etiology on peripheral endothelial function after cardiac transplantation is unknown. METHODS: Using brachial artery ultrasound, endothelium-dependent, flow-mediated dilation (FMD) was assessed in patients with heart failure with either nonischemic cardiomyopathy (n = 10) or ischemic cardiomyopathy (n = 7), cardiac transplant recipients with prior nonischemic cardiomyopathy (n = 10) or prior ischemic cardiomyopathy (n = 10) and normal controls (n = 10). RESULTS: Patients with heart failure with either ischemic cardiomyopathy or nonischemic cardiomyopathy had impaired FMD (3.6 +/- 1.0% and 5.1 +/- 1.2%, respectively, p = NS) compared with normal subjects (13.9 +/- 1.3%, p < 0.01 compared with either heart failure group). In transplant recipients with antecedent nonischemic cardiomyopathy, FMD was markedly higher than that of heart failure patients with nonischemic cardiomyopathy (13.0 +/- 2.4%, p < 0.001) and similar to that of normal subjects (p = NS). However, FMD remained impaired in transplant recipients with prior ischemic cardiomyopathy (5.5 +/- 1.5%, p = 0.001 compared with normal, p = 0.002 vs. transplant recipients with previous nonischemic cardiomyopathy). CONCLUSIONS: Peripheral vascular endothelial function is normal in cardiac transplant recipients with antecedent nonischemic cardiomyopathy, but remains impaired in those with prior ischemic cardiomyopathy. In contrast, endothelial function is uniformly abnormal for patients with heart failure, regardless of etiology. These findings indicate that cardiac transplantation corrects peripheral endothelial function for patients without ischemic heart disease, but not in those with prior atherosclerotic coronary disease.


Assuntos
Endotélio Vascular/fisiopatologia , Insuficiência Cardíaca/etiologia , Transplante de Coração/fisiologia , Complicações Pós-Operatórias/etiologia , Vasodilatação/fisiologia , Adulto , Artéria Braquial/fisiopatologia , Feminino , Insuficiência Cardíaca/fisiopatologia , Humanos , Masculino , Pessoa de Meia-Idade , Isquemia Miocárdica/complicações , Isquemia Miocárdica/fisiopatologia , Isquemia Miocárdica/cirurgia , Complicações Pós-Operatórias/fisiopatologia , Fatores de Risco
18.
FEBS Lett ; 377(2): 103-8, 1995 Dec 18.
Artigo em Inglês | MEDLINE | ID: mdl-8543029

RESUMO

In women, estrogen (E2) exerts a clinically relevant anti-atherogenic effect. The atheroprotective effects of E2 are mediated both by E2-induced changes in systemic factors and by direct effects of E2 on the blood vessel wall. In studies to characterize E2 signaling pathways in vascular smooth muscle cells (VSMC), we recently demonstrated that human VSMC express a functional estrogen receptor [1]. In the present study, we applied a reverse transcription/PCR-based strategy to identify isoforms of the E2 receptor in human VSMC. We now report that in addition to the classical E2 receptor, human VSMC derived from both mammary artery and saphenous vein express an estrogen receptor isoform containing an in-frame deletion of Exon 4 (ER delta 4). RNase protection assays confirm the presence of ER delta 4 message in VSMC and demonstrate it is nearly as abundant as the classical E2 receptor. Transient transfection experiments in VSMC and HeLa cells demonstrate that, in contrast to the classical 67 kDa nuclear-localized E2 receptor, ER delta 4: (a) is a 55 kDa protein that is widely distributed throughout the cell; (b) does not transactivate an E2 response element-driven reporter plasmid in response to E2; and (c) does not modulate transactivation of the ERE-reporter by the classical (wild type) estrogen receptor. Thus, human VSMC express an E2 receptor isoform that does not appear to alter gene transcription. The presence of a novel isoform of the E2 receptor may have important implications for studies of E2-mediated signaling in VSMC.


Assuntos
Músculo Liso Vascular/metabolismo , Receptores de Estrogênio/biossíntese , Sequência de Bases , Sítios de Ligação , Células Cultivadas , Primers do DNA , Feminino , Células HeLa , Humanos , Masculino , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , RNA Mensageiro/metabolismo , Receptores de Estrogênio/química , Receptores de Estrogênio/genética
19.
FEBS Lett ; 502(3): 103-8, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11583108

RESUMO

Physiological effects of estrogen on myocardium are mediated by two intracellular estrogen receptors, ERalpha and ERbeta, that regulate transcription of target genes through binding to specific DNA target sequences. To define the role of ERbeta in the transcriptional activation of both endothelial (eNOS) and inducible nitric oxide synthase (iNOS) in cardiac myocytes, we used the complete ER-specific antagonist R,R-tetrahydrochrysene (R,R-THC). R,R-THC inhibited activation of iNOS/eNOS promoter-luciferase reporter constructs (iNOS/eNOS-Luc) in a dose-dependent fashion in COS7 cells selectively transfected with ERbeta, but failed to influence ERalpha-mediated increase of iNOS/ eNOS-Luc. In neonatal rat cardiomyocytes transfected with eNOS-Luc or iNOS-Luc, incubation with 17betaestradiol (E2, 10(-8) M) for 24 h stimulated expression of eNOS and iNOS. R,R-THC (10(-5) M) completely inhibited this effect. Furthermore, eNOS and iNOS protein expression in cardiac myocytes induced by E2 was completely blocked by R,R-THC as shown by immunoblot analysis. Taken together, these results show that ERbeta mediates transcriptional activation of eNOS and iNOS by E2.


Assuntos
Estradiol/farmacologia , Miocárdio/enzimologia , Óxido Nítrico Sintase/metabolismo , Receptores de Estrogênio/metabolismo , Animais , Western Blotting , Células COS , Células Cultivadas , Crisenos/farmacologia , Receptor beta de Estrogênio , Coração/efeitos dos fármacos , Immunoblotting , Luciferases/metabolismo , Masculino , Miocárdio/química , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Regiões Promotoras Genéticas , Ratos , Ratos Wistar , Transfecção , Regulação para Cima
20.
Arch Ophthalmol ; 116(9): 1209-12, 1998 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9747681

RESUMO

OBJECTIVES: To investigate the concentrations of carboplatin and etoposide achieved in the aqueous and vitreous humors after intravenous infusion in nonhuman primates, and to investigate whether local administration of carboplatin might result in higher concentrations in the vitreous humor. METHODS: Macaca fascicularis primates were treated with 1 of 3 regimens: (1) intravenous carboplatin (18.7 mg/kg), etoposide (5 mg/kg), and vincristine sulfate (0.05 mg/kg), (2) peribulbar carboplatin (10 mg/mL), or (3) episcleral balloon carboplatin (10 mg/mL). Concentrations of chemotherapeutic agents were measured in the plasma and in the aqueous and vitreous humors. RESULTS: No measurable amount of etoposide was detected in the aqueous or vitreous humor after intravenous administration. Mean measured peak vitreous concentration of carboplatin after intravenous administration was 0.31 microg/mL, which was 1% of the peak plasma value. Mean measured peak vitreous concentrations of carboplatin after peribulbar or episcleral balloon administration were 2.38 microg/mL and 2.95 microg/mL, respectively, which represent 7.68- and 9.52-fold increases over the concentration achieved after intravenous administration. No serious toxic effect was observed in any animal. CONCLUSIONS: Peribulbar and episcleral balloon administration of carboplatin seemed to be safe and resulted in higher vitreous concentrations than intravenous administration in this model. These results suggest that these alternate routes of delivery should be explored in children with vitreous seeding of retinoblastoma.


Assuntos
Antineoplásicos/farmacocinética , Humor Aquoso/metabolismo , Carboplatina/farmacocinética , Etoposídeo/farmacocinética , Vincristina/farmacocinética , Corpo Vítreo/metabolismo , Absorção , Administração Tópica , Animais , Antineoplásicos/administração & dosagem , Disponibilidade Biológica , Carboplatina/administração & dosagem , Etoposídeo/administração & dosagem , Infusões Intravenosas , Injeções , Macaca fascicularis , Soluções Oftálmicas , Órbita , Distribuição Tecidual , Vincristina/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA