Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 136
Filtrar
1.
J Control Release ; 171(2): 234-40, 2013 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-23916883

RESUMO

Typically, inhaled drugs are rapidly absorbed into the bloodstream, which results in systemic side effects and a brief residence time in the lungs. PEGylation was evaluated as a novel strategy for prolonging the retention of small inhaled molecules in the pulmonary tissue. Hydrolysable ester conjugates of PEG1000, PEG2000, 2000, PEG3400 and prednisolone, a model drug cleared from the lungs within a few minutes, were synthesised and thoroughly characterised. The conjugates were stable in buffers with hydrolysis half-lives ranging from 1h to 70 h, depending on the pH and level of substitution. With the exception of PEG3400-prednisolone, conjugates did not induce a significant lactate dehydrogenase (LDH) release from Calu-3 cells after a 20 h exposure. Following nebulisation to isolated perfused rat lungs (IPRL), the PEG2000 and mPEG2000 conjugates reduced the maximum prednisolone concentration in the perfusate (Cmax) by 3.0 and 2.2 fold, respectively. Moreover, while prednisolone was undetectable in the perfusion solution beyond 20 min when the free drug was administered, prednisolone concentrations were still quantifiable after 40 min following delivery of the conjugates. This study is the first to demonstrate hydrolysable PEG drug ester conjugates are a promising approach for optimising the pharmacokinetic profile of small drugs delivered by inhalation.


Assuntos
Pulmão/metabolismo , Polietilenoglicóis/farmacocinética , Prednisolona/farmacocinética , Administração por Inalação , Animais , Linhagem Celular Tumoral , Preparações de Ação Retardada/administração & dosagem , Preparações de Ação Retardada/química , Preparações de Ação Retardada/farmacocinética , Ésteres , Humanos , Masculino , Modelos Biológicos , Peso Molecular , Polietilenoglicóis/administração & dosagem , Polietilenoglicóis/química , Prednisolona/administração & dosagem , Prednisolona/química , Ratos , Ratos Wistar
2.
J Viral Hepat ; 19(12): 872-80, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23121366

RESUMO

A new hepatitis B virus (HBV) protein, hepatitis B splice-generated protein (HBSP), has been detected in liver biopsy specimens from patients with chronic active hepatitis. The aim of this study was to characterize the phenotype and functions of peripheral HBSP-specific T cells and to determine whether these T-cell responses may be implicated in liver damage or viral control. Two groups of patients were studied: HBV-infected patients with chronic active hepatitis and HBV-infected patients who were inactive carriers of hepatitis B surface antigen. HBSP-specific T-cell responses were analysed ex vivo and after in vitro stimulation of peripheral blood mononuclear cells. Soluble cytokines and chemokines were analysed in sera and in cell culture supernatants. Few HBSP- or capsid-specific T-cell responses were detected in patients with chronic active hepatitis whereas frequency of HBV-specific T cells was significantly higher in inactive carrier patients. HBSP activated CD8+ and CD4+ T cells that recognized multiple epitopes and secreted inflammatory cytokines. The IL-12 level was significantly lower in sera from asymptomatic carrier patients compared to patients with chronic active hepatitis. IL-12 and IP-10 levels in the sera were significantly and independently correlated with both alanine amino transferase and HBV DNA levels. Our results show that the HBSP protein activates cellular immune responses in HBV-infected patients but has probably no prominent role in liver damage. The pattern of cytokines and chemokines in sera was linked to HBV viral load and was consistent with the level of inflammation during chronic hepatitis.


Assuntos
Citocinas/metabolismo , Vírus da Hepatite B/imunologia , Hepatite B Crônica/imunologia , Fígado/patologia , Linfócitos T/imunologia , Proteínas Virais/imunologia , Adulto , Idoso , Alanina Transaminase/sangue , Portador Sadio/imunologia , Portador Sadio/virologia , Células Cultivadas , Citocinas/sangue , Feminino , Hepatite B Crônica/virologia , Humanos , Leucócitos Mononucleares/imunologia , Fígado/virologia , Masculino , Pessoa de Meia-Idade , Subpopulações de Linfócitos T/imunologia , Carga Viral , Adulto Jovem
3.
Clin Exp Pharmacol Physiol ; 35(4): 396-401, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18307728

RESUMO

1. Although hormonal therapy (HT) may increase the risk of coronary heart disease (CHD) and stroke in postmenopausal women, epidemiological studies (protection in premenopausal women) suggest and experimental studies (prevention of fatty streak development in animals) demonstrate a major atheroprotective action of estradiol (E2). The understanding of the deleterious and beneficial effects of oestrogens is thus required. 2. The immuno-inflammatory system plays a key role in the development of fatty streak deposit as well as in the rupture of the atherosclerotic plaque. Although E2 favours an anti-inflammatory effect in vitro (cultured cells), it rather elicits a pro-inflammatory response in vivo involving several subpopulations of the immuno-inflammatory system, which could contribute to plaque destabilization. The functional role of several cytokines was explored in hypercholesterolemic mice. The atheroprotective effect of E2 was fully maintained in mice deficient in interferon-g or interleukin-12, as well as IL-10. In contrast, the protective effect of estradiol was abolished and even reversed in hypercholesterolemic mice given a neutralizing anti-transforming growth factor-b (TGF-b) antibody. Endothelium is another important target for E2, since it not only potentiates endothelial nitric oxide and prostacyclin production, but also controls trafficking of the populations of the immuno-inflammatory system. 3. To conclude, the respective actions of oestrogens on the cell populations involved in the pathophysiology of atherothrombosis may be influenced, among others, by the timing of HT initiation, the status of the vessel wall and, as recently demonstrated the status of the TGF-b pathway.


Assuntos
Aterosclerose/metabolismo , Citocinas/metabolismo , Estradiol/metabolismo , Estradiol/farmacologia , Animais , Endotélio/metabolismo , Feminino , Deleção de Genes , Humanos , Hipercolesterolemia , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Camundongos , Fator de Crescimento Transformador beta
4.
Arch Mal Coeur Vaiss ; 100(6-7): 554-62, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17893638

RESUMO

Whereas hormonal replacement/menopause therapy (HRT) in post-menopausal women increases the coronary artery risk, epidemiological studies (protection in pre-menopaused women) suggest and experimental studies (prevention of the development of fatty streaks in animals) demonstrate a major atheroprotective action of estradiol (E2). The understanding of the deleterious and beneficial effects of estrogens is thus required. The immuno-inflammatory system plays a key role in the development of fatty streak deposit as well as in the atherosclerotic plaque rupture. Whereas E2 favors an anti-inflammatory effect in vitro (cultured cells), it rather elicits pro-inflammation in vivo, at the level of several subpopulations of the immuno-inflammatory system, which could contribute to plaque destabilization. Endothelium is another important target for E2, since it stimulates endothelial NO and prostacyclin production, thus promoting beneficial effects of vasorelaxation and platelet aggregation inhibition. Prostacyclin, but not NO, appears to be involved in the atheroprotective effect of E2. Estradiol accelerates also endothelial regrowth, thus favoring vascular healing. Finally, most of these effects of E2 are mediated by estrogen receptor alpha, and are independent of estrogen receptor beta. In summary, a better understanding of the mechanisms of estrogen action is required not only on the normal and atheromatous arteries, but also on innate and adaptive immune responses. This should help cardiovascular disease prevention optimization after menopause. These mouse models should help to screen existing and future Selective Estrogen Receptor Modulators (SERMs).


Assuntos
Estradiol/farmacologia , Terapia de Reposição de Estrogênios , Animais , Anti-Inflamatórios/farmacologia , Doença da Artéria Coronariana/induzido quimicamente , Doença da Artéria Coronariana/prevenção & controle , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Estradiol/efeitos adversos , Feminino , Humanos , Mediadores da Inflamação/farmacologia , Camundongos , Pós-Menopausa/efeitos dos fármacos , Pré-Menopausa/efeitos dos fármacos , Fatores de Risco , Moduladores Seletivos de Receptor Estrogênico/farmacologia
6.
Circ Res ; 94(10): 1301-9, 2004 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-15073041

RESUMO

Both 17beta-estradiol (E2) and fibroblast growth factor-2 (FGF2) stimulate angiogenesis and endothelial cell migration and proliferation. The first goal of this study was to explore the potential link between this hormone and this growth factor. E2-stimulated angiogenesis in SC Matrigel plugs in Fgf2+/+ mice, but not in Fgf2-/- mice. Cell cultures from subcutaneous Matrigel plugs demonstrated that E2 increased both migration and proliferation in endothelial cells from Fgf2+/+ mice, but not from in Fgf2-/- mice. Several isoforms of fibroblast growth factor-2 (FGF2) are expressed: the low molecular weight 18-kDa protein (FGF2lmw) is secreted and activates tyrosine kinase receptors (FGFRs), whereas the high molecular weight (21 and 22 kDa) isoforms (FGF2hmw) remains intranuclear, but their role is mainly unknown. The second goal of this study was to explore the respective roles of FGF2 isoforms in the effects of E2. We thus generated mice deficient only in the FGF2lmw (Fgf2lmw-/-). E2 stimulated in vivo angiogenesis and in vitro migration in endothelial cells from Fgf2lmw-/- as it did in Fgf2+/+ mice. E2 increased FGF2hmw protein abundance in endothelial cell cultures from Fgf2+/+ and Fgf2lmw-/- mice. As shown using siRNA transfection, these effects were FGFR independent but involved FGF2-Interacting Factor, an intracellular FGF2hmw partner. This is the first report for a physiological role for the intracellular FGF2hmw found to mediate the effect of E2 on endothelial cell migration via an intracrine action.


Assuntos
Endotélio Vascular/fisiologia , Estradiol/farmacologia , Fator 2 de Crescimento de Fibroblastos/fisiologia , Neovascularização Fisiológica , Animais , Divisão Celular , Movimento Celular , Células Cultivadas , Endotélio Vascular/citologia , Receptor alfa de Estrogênio , Fator 2 de Crescimento de Fibroblastos/genética , Camundongos , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Receptores de Estrogênio/fisiologia , Receptores de Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais
7.
Eur J Endocrinol ; 150(2): 113-7, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14763907

RESUMO

Numerous epidemiological as well as experimental studies have suggested that estradiol (E2) prevents atherosclerosis development. However two controlled prospective and randomized studies in women using hormone replacement therapy (HRT) did not confirm this beneficial effect. We then decided to use mouse models of atherosclerosis to define the possible mechanisms involved and the reasons for the discrepancy. We have shown that, although serum cholesterol decreases, this influence on lipid metabolism is negligible. Surprisingly, E2 induces an inflammatory-immune response towards a T helper cell (Th1) profile with increasing interferon-gamma production that could destabilize atheromatous plaques, and could account for the increase in the frequency of cardiovascular events in women undergoing HRT. At the level of the endothelium, E2 induces an increase in nitric oxide (NO) biodisponibility, but this phenomenon does not concern the development of fatty streaks. Nevertheless, the atheroprotective effect is apparently mediated at the level of the endothelium by a mechanism that has still to be characterized in molecular terms. These new acquisitions constitute a basis for new pharmacological developments allowing the prevention of deleterious effects and preserving the beneficial ones.


Assuntos
Arteriosclerose/fisiopatologia , Endotélio Vascular/fisiopatologia , Estradiol/metabolismo , Animais , Arteriosclerose/imunologia , Modelos Animais de Doenças , Feminino , Humanos , Inflamação/fisiopatologia , Camundongos , Camundongos Knockout , Células Th1/fisiologia
8.
Atherosclerosis ; 166(1): 41-8, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12482549

RESUMO

The mechanisms that mediate the atheroprotective properties of estrogens remain obscure. In the present study, we evaluated the involvement of the adhesion molecule P-selectin, intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1) in the atheroprotective effect of estrogens in murine models evaluating early steps of atherosclerosis. First, we studied the effect of 17 beta-estradiol (E2) administration for 12 weeks on fatty streak constitution at the root aorta of ovariectomized female mice deficient in apolipoprotein E (apoE) alone or deficient in both apoE and either P-selectin or ICAM-1. Compared with respective placebo groups, E2 significantly prevented the development of fatty streak, to a similar extent in all three genotypes (-70.0% in apoE(-/-), -77.4% in apoE(-/-) P-selectin(-/-), and -77.1% in apoE(-/-) ICAM-1(-/-)). Second, the endothelial expression of VCAM-1 at the root aorta was assessed by immunohistochemistry in either placebo or E2-treated ovariectomized C57BL/6 female mice fed an atherogenic diet. Compared with placebo, E2 treatment resulted in a 31.8% decrease of VCAM-1 endothelial expression at this lesion-prone site (P=0.03). These results demonstrate that P-selectin and ICAM-1 are not involved in the atheroprotective effect of estrogens, and suggest that VCAM-1 could play a role in this process.


Assuntos
Apolipoproteínas E/deficiência , Arteriosclerose/metabolismo , Estradiol/farmacologia , Molécula 1 de Adesão Intercelular/metabolismo , Selectina-P/metabolismo , Molécula 1 de Adesão de Célula Vascular/metabolismo , Animais , Apolipoproteínas E/genética , Arteriosclerose/patologia , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Molécula 1 de Adesão de Célula Vascular/efeitos dos fármacos
10.
Circ Res ; 90(4): 413-9, 2002 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-11884370

RESUMO

Although estradiol (E(2)) has been recognized to exert several vasculoprotective effects in several species, its effects in mouse vasomotion are unknown, and consequently, so is the estrogen receptor subtype mediating these effects. We investigated the effect of E(2) (80 microg/kg/day for 15 days) on NO production in the thoracic aorta of ovariectomized C57Bl/6 mice compared with those given placebo. E(2) increased basal NO production. In contrast, the relaxation in response to ATP, to the calcium ionophore A23187, and to sodium nitroprusside was unaltered by E(2), whereas acetylcholine-elicited relaxation was decreased. The abundance of NO synthase I, II, and III immunoreactive proteins (using Western blot) in thoracic aorta homogenates was unchanged by E(2). To determine the estrogen receptor (ER) subtype involved in these effects, transgenic mice in which either the ERalpha or ERbeta has been disrupted were ovariectomized and treated, or not, with E(2). Basal NO production was increased and the sensitivity to acetylcholine decreased in ERbeta knockout mice in response to E(2), whereas this effect was abolished in ERalpha knockout mice. Finally, these effects of E(2) on vasomotion required long-term and/or in vivo exposure, as short-term incubation of aortic rings with 10 nmol/L E(2) in the isolated organ chamber did not elicit any vasoactive effects. In conclusion, this study demonstrates that ERalpha, but not ERbeta, mediates the beneficial effect of E(2) on basal NO production.


Assuntos
Aorta Torácica/efeitos dos fármacos , Aorta Torácica/metabolismo , Estradiol/administração & dosagem , Óxido Nítrico/metabolismo , Receptores de Estrogênio/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Fatores Biológicos/metabolismo , Peso Corporal/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase/farmacologia , Relação Dose-Resposta a Droga , Implantes de Medicamento , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Feminino , Técnicas In Vitro , Ionóforos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico Sintase/metabolismo , Nitroprussiato/farmacologia , Ovariectomia , Receptores de Estrogênio/deficiência , Receptores de Estrogênio/genética , Útero/efeitos dos fármacos , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia
11.
Proc Natl Acad Sci U S A ; 99(4): 2205-10, 2002 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-11854517

RESUMO

Two isoforms of estrogen receptor (ER) have been described: ERalpha and ERbeta. The initial gene targeting of ERalpha, consisting in the introduction of a Neo cassette in exon 1 [alphaERKO, hereafter called ERalpha-Neo KO (knockout)], was reported in 1993. More recently, another mouse deficient in ERalpha because of the deletion of exon 2 (ERalphaKO, hereafter called ERalpha-delta2 KO) was generated. In ovariectomized ERalpha-wild-type mice, estradiol (E(2)) increases uterine weight and basal production of endothelial nitric oxide (NO). Both of these effects are abolished in ERalpha-delta2 KO mice. In contrast, we show here that both of these effects of E(2) are partially (uterine weight) or totally (endothelial NO production) preserved in ERalpha-Neo KO. We also confirm the presence of two ERalpha mRNA splice variants in uterus and aorta from ERalpha-Neo KO mice. One of them encodes a chimeric ERalpha protein (ERalpha55), partially deleted in the A/B domain, that was detected in both uterus and aorta by Western blot analysis. The other ERalpha mRNA splice variant codes for an isoform deleted for the A/B domain (ERalpha46), which was detected in uterus of ERalpha-Neo KO, and wild-type mice. This protein isoform was not detected in aorta. The identification of these two N-terminal modified isoforms in uterus, and at least one of them in aorta, probably explains the persistence of the E(2) effects in ERalpha-Neo KO mice. Furthermore, ERalpha-Neo KO mice may help in the elucidation of the specific functions of full-length ERalpha (ERalpha66) and ERalpha46, both shown to be physiologically generated in vivo.


Assuntos
Estradiol/farmacologia , Óxido Nítrico/biossíntese , Receptores de Estrogênio/genética , Receptores de Estrogênio/fisiologia , Processamento Alternativo , Animais , Aorta/metabolismo , Western Blotting , Relação Dose-Resposta a Droga , Endotélio Vascular/metabolismo , Receptor alfa de Estrogênio , Éxons , Feminino , Hipertrofia , Imuno-Histoquímica , Camundongos , Camundongos Knockout , Modelos Genéticos , Mutagênese Insercional , Tamanho do Órgão , Isoformas de Proteínas , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Útero/efeitos dos fármacos , Útero/metabolismo , Útero/patologia
12.
J Am Chem Soc ; 123(15): 3520-40, 2001 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-11472124

RESUMO

The reactions of CpZr(CH(3))(3), 1, and Cp(2)Zr(CH(3))(2), 2, with partially dehydroxylated silica, silica-alumina, and alumina surfaces have been carried out with careful identification of the resulting surface organometallic complexes in order to probe the relationship between catalyst structure and polymerization activity. The characterization of the supported complexes has been achieved in most cases by in situ infrared spectroscopy, surface microanalysis, qualitative and quantitative analysis of evolved gases during surface reactions with labeled surface, solid state (1)H and (13)C NMR using (13)C-enriched compounds, and EXAFS. 1 and 2 react with silica(500) and silica-alumina(500) by simple protonolysis of one Zr-Me bond by surface silanols with formation of a single well-defined neutral compound. In the case of silica-alumina, a fraction of the supported complexes exhibits some interactions with electronically unsaturated surface aluminum sites. 1 and 2 also react with the hydroxyl groups of gamma-alumina(500), leading to several surface structures. Correlation between EXAFS and (13)C NMR data suggests, in short, two main surface structures having different environments for the methyl group: [Al](3)-OZrCp(CH(3))(2) and [Al](2)-OZrCp(CH(3))(mu-CH(3))-[Al] for the monoCp series and [Al](2)-OZrCp(2)(CH(3)) and [Al]-OZrCp(2)(mu-CH(3))-[Al] for the bisCp series. Ethylene polymerization has been carried out with all the supported complexes under various reaction conditions. Silica-supported catalysts in the absence of any cocatalyst exhibited no activity whatsoever for ethylene polymerization. When the oxide contained Lewis acidic sites, the resulting surface species were active. The activity, although improved by the presence of additional cocatalysts, remained very low by comparison with that of the homogeneous metallocene systems. This trend has been interpreted on the basis of various possible parameters, including the (p-pi)-(d-pi) back-donation of surface oxygen atoms to the zirconium center.

13.
Atherosclerosis ; 156(2): 315-20, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11395027

RESUMO

Interleukin-6 (IL-6) gene expressed in bone marrow-derived stromal cells and osteoblasts contributes to the state of mineralization and its control by estradiol may be involved in the development of post-menopausal osteoporosis. Since IL-6 is also expressed in the different cell populations of the arterial wall, the purpose of this study was to gain more insight into its involvement in the atherosclerotic process and the atheroprotective effect of estradiol by studying double deficient mice at the apolipoprotein E and IL-6 loci (IL-6(-/-)/E(-/-)). At 1 year of age, IL-6(-/-)/E(-/-) mice showed similar hypercholesterolemia to IL-6(+/+)/E(-/-) mice but presented significantly larger and more calcified lesions. In younger mice (sixteen weeks of age), no significant difference in fatty streaks could be detected in IL-6(+/+)/E(-/-), IL-6(+/-)/E(-/-) and IL-6(-/-)/E(-/-) mice on a normal chow diet. Estrogen supplementation at this age induced a decrease of fatty streak formation in all three genotypes. The combined data indicate that IL-6 expression is involved at the fibrous plaque stage of the atherosclerotic process but does not constitute a direct target for estradiol to prevent fatty streak formation.


Assuntos
Apolipoproteínas E/deficiência , Arteriosclerose/patologia , Estradiol/metabolismo , Interleucina-6/deficiência , Interleucina-6/metabolismo , Seio Aórtico/patologia , Análise de Variância , Animais , Arteriosclerose/tratamento farmacológico , Técnicas de Cultura , Modelos Animais de Doenças , Estradiol/uso terapêutico , Feminino , Interleucina-6/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fotomicrografia , Probabilidade , Sensibilidade e Especificidade , Especificidade da Espécie
14.
Atherosclerosis ; 155(2): 291-5, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11254898

RESUMO

Angiotensin-converting enzyme (ACE) is mainly responsible for converting angiotensin I (AI) to angiotensin II (AII), and ACE inhibitors prevent atherosclerosis in animal models. Neutral endopeptidase 24.11 (NEP) degrades substance P, kinins and atrial natriuretic peptide (ANP), and aortic wall NEP activity was found to be increased in atherosclerosis. In the present study, we have evaluated the effect of candoxatril, a NEP inhibitor, and of omapatrilat, a dual ACE and NEP inhibitor, on the development of fatty streak in apolipoprotein E (apoE)-deficient mice. Groups of ten male apoE-deficient mice were given either placebo, candoxatril 50 mg/kg per day, or omapatrilat 10, or 100 mg/kg per day for 4 months. None of the treatments influenced body weight, serum total or HDL-cholesterol. Compared with the placebo, candoxatril did not protect the mice from fatty streak deposit. In contrast, omapatrilat dose dependently inhibited the constitution of fatty streak in apoE-deficient mice. The precise advantages of the dual ACE and NEP inhibition versus the inhibition of only ACE should now be considered in the prevention of atherosclerosis as well as in the occurrence of its complications.


Assuntos
Inibidores da Enzima Conversora de Angiotensina/uso terapêutico , Apolipoproteínas E/deficiência , Arteriosclerose/prevenção & controle , Neprilisina/antagonistas & inibidores , Inibidores de Proteases/uso terapêutico , Piridinas/uso terapêutico , Tiazepinas/uso terapêutico , Inibidores da Enzima Conversora de Angiotensina/farmacologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Arteriosclerose/enzimologia , Fator Natriurético Atrial/metabolismo , Peso Corporal/efeitos dos fármacos , Bradicinina/metabolismo , Colesterol/sangue , HDL-Colesterol/sangue , Avaliação Pré-Clínica de Medicamentos , Indanos/farmacologia , Indanos/uso terapêutico , Masculino , Camundongos , Camundongos Knockout , Propionatos/farmacologia , Propionatos/uso terapêutico , Inibidores de Proteases/farmacologia , Piridinas/farmacologia , Substância P/metabolismo , Tiazepinas/farmacologia , Triglicerídeos/sangue
15.
Circulation ; 103(3): 423-8, 2001 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-11157695

RESUMO

BACKGROUND: The atheroprotective effect of 17beta-estradiol (E(2)) has been suggested in women and clearly demonstrated in animals through both an effect on lipid metabolism and a direct effect on the cells of the arterial wall. It has been shown, for example, that E(2) promotes endothelium-dependent relaxation and accelerates reendothelialization in rats. Similar studies have been undertaken in mice to appreciate the molecular mechanism of this process. METHODS AND RESULTS: We report here a model of electric carotid injury adapted from that described by Carmeliet et al (1997) that allows us to precisely evaluate the reendothelialization process. We demonstrate that E(2) accelerates endothelial regeneration in castrated female wild-type mice. In ovariectomized transgenic mice in which either the estrogen receptor (ER)-alpha or ERbeta gene has been disrupted, E(2) accelerated reendothelialization in female ERbeta knockout mice, whereas this effect was abolished in female ERalpha knockout mice. CONCLUSIONS: This study demonstrates that ERalpha but not ERbeta mediates the beneficial effect of E(2) on reendothelialization and potentially the prevention of atherosclerosis.


Assuntos
Lesões das Artérias Carótidas/tratamento farmacológico , Artéria Carótida Primitiva/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Estradiol/farmacologia , Receptores de Estrogênio/efeitos dos fármacos , Animais , Arteriosclerose/prevenção & controle , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/fisiopatologia , Artéria Carótida Primitiva/metabolismo , Artéria Carótida Primitiva/ultraestrutura , Castração , Modelos Animais de Doenças , Estradiol/sangue , Receptor alfa de Estrogênio , Receptor beta de Estrogênio , Azul Evans , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Varredura , Receptores de Estrogênio/deficiência , Regeneração , Coloração e Rotulagem , Fatores de Tempo
16.
Clin Exp Pharmacol Physiol ; 28(12): 1032-4, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11903309

RESUMO

1. The rationale for preclinical research on the atheroprotective effect of oestrogens is based on the epidemiological evidence that women are protected against the clinical complications of atherosclerosis until menopause. However, this protection, probably due to sex hormones, is progressively lost within the years following menopause. 2. In addition, numerous studies have clearly demonstrated the atheroprotective effect of oestrogens in all animal models. 3. In the present paper, we first summarize our understanding of the pathophysiology of atherosclerosis. We then focus on the recognized target of oestradiol (E2) in the vessel wall: the classical target, namely the endothelium, and a recently characterized target, namely cells of the inflammatory-immune system. Finally, we discuss how unknown mechanisms in atherosclerosis could be responsible for the absence of effect of hormone-replacement therapy in the Heart and Estrogen/ progestin Replacement Study (HERS).


Assuntos
Arteriosclerose/fisiopatologia , Endotélio Vascular/efeitos dos fármacos , Estrogênios/farmacologia , Arteriosclerose/tratamento farmacológico , Endotélio Vascular/fisiologia , Terapia de Reposição de Estrogênios , Estrogênios/uso terapêutico , Feminino , Humanos , Pós-Menopausa/fisiologia
17.
Cytokine ; 12(7): 1110-4, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10880259

RESUMO

Fusion proteins consisting of the 24 kDa nuclear form of basic fibroblast growth factor (FGF-2), associated with the hormone binding domain of oestrogen receptor (HBD), convey oestrogen inducibility to FGF-2. When stable HBD-FGF-2 HeLa cell lines were transiently transfected with an interleukin 6 (IL-6) construct, the IL-6 promoter activity was downregulated by the addition of oestradiol. Moreover, in these cell lines, the function of the FGF-2 nuclear localisation sequence was abolished by its fusion to HBD, while addition of oestradiol restored the location of the chimera to the nucleus.


Assuntos
Regulação para Baixo , Estrogênios/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Interleucina-6/genética , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Sítios de Ligação , Western Blotting , Estradiol/metabolismo , Estradiol/farmacologia , Fator 2 de Crescimento de Fibroblastos/genética , Imunofluorescência , Células HeLa , Humanos , Ligação Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Receptores de Estrogênio/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
18.
J Vasc Res ; 37(3): 202-8, 2000.
Artigo em Inglês | MEDLINE | ID: mdl-10859479

RESUMO

In addition to their actions on reproductive function, estrogens have important effects on endothelial cells. The present study was designed to evaluate the mechanism(s) by which 17beta-estradiol (E2) promotes endothelial cell proliferation. The potential involvement of vascular endothelial growth factor (VEGF) was investigated by the coadministration of polyclonal anti-VEGF antibody. First, the effect of E2 on the proliferation of cultured foetal bovine aortic endothelial cells (FBAEC) was studied. E2 stimulated this proliferation with an EC50 between 10(-11) and 10(-10) M and this effect was inhibited by the anti-VEGF antibody. The effect of a physiological dose of E2 was then studied in the rat model of carotid injury. After deendothelializing balloon injury, reendothelialization of the denuded surface may influence the growth of the underlying smooth muscle cells. Male Sprague-Dawley rats were castrated and then received E2 from subcutaneously implanted pellets that released 3.2 microg/kg/day. Endothelial regrowth (Evans blue staining) and neointimal thickening were evaluated 2 weeks after the carotid injury. In comparison to the placebo group, E2 increased the extent of reendothelialization (p = 0.0002) and reduced neointimal thickening (p = 0.0007). Anti-VEGF antibody abolished the effect of E2 on reendothelialization as well as on neointimal thickening. Thoracic aorta VEGF content was increased in E2-treated rats compared to control rats. In conclusion, the present study demonstrates that E2 increases endothelial cell proliferation in vitro and reendothelialization in vivo by means of a mechanism dependent on endogenous VEGF. This effect could contribute to the antiatherogenic effect of a physiological dose of E2.


Assuntos
Fatores de Crescimento Endotelial/fisiologia , Endotélio Vascular/patologia , Endotélio Vascular/fisiopatologia , Estradiol/farmacologia , Linfocinas/fisiologia , Mitógenos/farmacologia , Cicatrização/fisiologia , Animais , Aorta Torácica/embriologia , Aorta Torácica/lesões , Aorta Torácica/patologia , Aorta Torácica/fisiopatologia , Cateterismo/efeitos adversos , Bovinos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Fatores de Crescimento Endotelial/biossíntese , Endotélio Vascular/embriologia , Endotélio Vascular/lesões , Feto , Linfocinas/biossíntese , Masculino , Ratos , Ratos Sprague-Dawley , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
19.
Ann Endocrinol (Paris) ; 61(1): 80-4, 2000 Feb.
Artigo em Francês | MEDLINE | ID: mdl-10790597

RESUMO

Two isoforms of oestrogens receptor (alpha and B) have been identified in the cells of the arterial wall, and an heterogenity of their expression according to the animal species, to the vascular bed and to sex has been reported. Estrogens can thus directly influence the vascular physiology through a genomic mechanism, but extra-genomic mechanisms responsible for a short-term effect have also been suggested. Endothelium appears to be an important target for estradiol, because this hormone potentiates endothelium-dependant relaxation through an increase in NO bioavailability, and accelerates endothelial regrowth. In the model of apolipoprotein E-deficient mice, as the atrhroprotective effect deposit. The immune system appears to play a key role, as the athroprotective effect of estradiol is absent in mice deficient in T and B lymphocytes. Estrogens potentiate the endothelium-dependant relaxation through the increase in nitric oxide bioavailability. Endothelial dysfunction (abnormality of the endothelium-dependent vasodilation) occurs in atheromatous arteries. Estrogens prevent and even correct this endothelial dysfunction. In monkeys, this beneficial effect of estrogens is not altered by coadministration of progesterone, but is abolished.


Assuntos
Artérias/fisiopatologia , Estrogênios/fisiologia , Animais , Artérias/efeitos dos fármacos , Arteriosclerose/fisiopatologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/fisiopatologia , Estradiol/farmacologia , Humanos , Relaxamento Muscular/efeitos dos fármacos , Músculo Liso Vascular/fisiopatologia , Receptores de Estrogênio/fisiologia
20.
Endocrinology ; 141(1): 462-5, 2000 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-10614672

RESUMO

Estradiol significantly decreases fatty streak formation in the aortic root of chow-fed apolipoprotein E-deficient mice. In contrast, immunodeficient mice with homozygous disruption at the recombinase activating gene 2 loci present fatty streak development that is insensitive to estradiol. Lymphocytes thus appear to be required for development of the atheroprotective effect of estradiol in this mouse model.


Assuntos
Apolipoproteínas E/genética , Arteriosclerose/tratamento farmacológico , Estradiol/uso terapêutico , Animais , Aorta/patologia , Apolipoproteínas E/imunologia , Arteriosclerose/genética , Arteriosclerose/imunologia , Arteriosclerose/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Feminino , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Transposases/genética , Transposases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA