Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Gene Ther ; 15(1): 1-11, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17637794

RESUMEN

A single plasmid regulated expression vector based upon a mifepristone-inducible two plasmid system, termed pBRES, has been constructed and tested in mice using murine interferon-b (mIFNb) as the transgene. The expression of mIFNb in the circulation was followed by measuring the systemic induction of IP-10, a validated biomarker for mIFNb in mice. Long-term, inducible expression of mIFNb was demonstrated following a single intramuscular (i.m.) injection of the pBRES mIFNb plasmid vector into the hind limb of mice. Induction of mIFNb expression was achieved by administration of the small molecule inducer, mifepristone (MFP). Plasmid DNA and mIFNb mRNA levels in the injected muscles correlated with mIFNb expression as monitored by IP-10 over a 3-month time period. Renewable transgene expression was achieved following repeat administration of the plasmid at 3 months following the first plasmid injection. A dose-dependent increase in expression was demonstrated by varying the amount of injected plasmid or the amount of the inducer administered to the mice. Finally, the pBRES plasmid expressing mIFNb under control of the inducer, MFP, was shown to be efficacious in a murine model of experimental allergic encephalomyelitis, supporting the feasibility of gene-based therapeutic approaches for treating diseases such as multiple sclerosis.


Asunto(s)
Encefalomielitis Autoinmune Experimental/terapia , Regulación de la Expresión Génica , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Interferón beta/genética , Plásmidos/administración & dosificación , Animales , Biomarcadores/sangre , Quimiocina CXCL10/análisis , Progresión de la Enfermedad , Femenino , Inyecciones Intramusculares , Interferón beta/sangre , Ratones , Ratones Endogámicos , Mifepristona/administración & dosificación , Esclerosis Múltiple/terapia , Plásmidos/análisis , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transgenes
2.
J Clin Invest ; 99(10): 2429-37, 1997 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-9153286

RESUMEN

The present study was designed to test the hypothesis that estrogen receptors (ER) in the blood vessel wall play a role in the modulation of the release of endothelium-derived nitric oxide (EDNO). Both basal and stimulated release of EDNO were determined in aortic rings isolated from female and male wild-type and male homozygous estrogen receptor knock-out (ERKO) mice. 125I-17beta-estradiol binding in aortic tissue showed significantly more high affinity cytosolic- nuclear-binding sites in male compared with female wildtype mice. Estrogen receptor transcripts were present in the aorta of male wild-type mice, but they were absent in male ERKO animals. Basal release of EDNO (determined by endothelium-dependent contraction caused by NG-nitro-arginine) was significantly higher in aorta of wild-type male mice compared with wild-type female mice, and significantly lower in the aorta of male ERKO compared with male wild-type mice. Acetylcholine-induced endothelium-dependent relaxation was similar in all groups studied. No difference was observed in the activity of calcium-dependent nitric oxide synthase in homogenates of lungs and brain taken from male wild-type and ERKO mice. These studies show a significant association between the number of estrogen receptors and basal release of EDNO in the aorta of mice, and suggest that decreased vascular estrogen receptor number may represent a novel risk factor for cardiovascular diseases.


Asunto(s)
Endotelio Vascular/fisiología , Músculo Liso Vascular/fisiología , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico/biosíntesis , Nitroarginina/farmacología , Receptores de Estrógenos/biosíntesis , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Animales , Aorta/fisiología , Encéfalo/enzimología , Cartilla de ADN , Endotelio Vascular/efectos de los fármacos , Inducción Enzimática , Estradiol/sangre , Exones , Femenino , Técnicas In Vitro , Pulmón/enzimología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Reacción en Cadena de la Polimerasa , Endoperóxidos de Prostaglandinas Sintéticos/farmacología , Receptores de Estrógenos/genética , Caracteres Sexuales , Especificidad de la Especie , Tromboxano A2/análogos & derivados , Tromboxano A2/farmacología , Vasoconstrictores/farmacología
3.
Hum Gene Ther ; 16(10): 1202-11, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16218781

RESUMEN

The objectives of this study were to assess the safety and potential clinical efficacy of adenovirus-delivered fibroblast growth factor-4 (Ad5FGF-4) by intramuscular injection into patients with critical limb ischemia (CLI). This study was a double-blind, randomized, placebo-controlled study with escalating dose groups of 2.87 x 10(8) to 2.87 x 10(10) viral particles. Thirteen patients with CLI were randomized to receive active drug (n = 10) or placebo (n = 3). Safety evaluations and efficacy parameters (ankle-brachial index, digital subtraction angiograms, magnetic resonance imaging, and scintigraphy) were performed at baseline and for 12 weeks after treatment. Injections of Ad5FGF-4 were generally well tolerated and considered safe. Transfection efficacy at these concentrations may have been limited or local. The small sample size did not allow any firm conclusions regarding clinical efficacy but a trend toward more and slightly larger blood vessels was observed in the angiograms. It is concluded that intramuscular injection of Ad5FGF-4 into CLI patients seemed safe, but transfection efficacy was limited at the assessed doses. Conclusions regarding clinical efficacy are impossible to draw from this small patient cohort.


Asunto(s)
Adenoviridae , Extremidades/irrigación sanguínea , Terapia Genética , Vectores Genéticos/administración & dosificación , Isquemia/terapia , Anciano , Anciano de 80 o más Años , Estudios de Cohortes , Método Doble Ciego , Femenino , Terapia Genética/métodos , Humanos , Inyecciones Intramusculares , Isquemia/metabolismo , Masculino , Persona de Mediana Edad
4.
J Gen Physiol ; 110(2): 135-40, 1997 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9236206

RESUMEN

Steroid hormones control the expression of many cellular regulators, and a role for estrogen in cardiovascular function and disease has been well documented. To address whether the activity of the L-type Ca2+ channel, a critical element in cardiac excitability and contractility, is altered by estrogen and its nuclear receptor, we examined cardiac myocytes from male mice in which the estrogen receptor gene had been disrupted (ERKO mice). Binding of dihydropyridine Ca2+ channel antagonist isradipine (PN200-110) was increased 45.6% in cardiac membranes from the ERKO mice compared to controls, suggesting that a lack of estrogen receptors in the heart increased the number of Ca2+ channels. Whole-cell patch clamp of acutely dissociated adult cardiac ventricular myocytes indicated that Ca2+ channel current was increased by 49% and action potential duration was increased by 75%. Examination of electrocardiogram parameters in ERKO mice showed a 70% increase in the QT interval without significant changes in PQ or QRS intervals. These results show that the membrane density of the cardiac L-type Ca2+ channel is regulated by the estrogen receptor and suggest that decreased estrogen may lead to an increase in the number of cardiac L-type Ca2+ channels, abnormalities in cardiac excitability, and increased risk of arrhythmia and cardiovascular disease.


Asunto(s)
Canales de Calcio/metabolismo , Miocardio/metabolismo , Receptores de Estrógenos/genética , Potenciales de Acción/efectos de los fármacos , Potenciales de Acción/fisiología , Animales , Bloqueadores de los Canales de Calcio/farmacología , Dihidropiridinas/metabolismo , Estimulación Eléctrica , Electrofisiología , Isradipino/farmacología , Ligandos , Masculino , Ratones , Ratones Noqueados , Miocardio/citología , Técnicas de Placa-Clamp , Receptores de Estrógenos/fisiología , Factores de Transcripción/fisiología
5.
Cardiovasc Res ; 50(1): 108-14, 2001 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11282083

RESUMEN

OBJECTIVE: The cellular basis of the cardioprotective effects of estrogen are largely unknown. An inhibitory effect on vascular smooth muscle (VSM) growth has been proposed. We examined the effect of 17beta-estradiol (E2) on mechanical strain-induced mitogenesis in human fetal VSM cells. METHODS AND RESULTS: Cells were grown on fibronectin-coated plates with silicone-elastomer bottoms, and exposed to cyclic mechanical strain (60 cycles/min), with and without E2 (1 nmol/l), for 48 h. [3H]-Thymidine incorporation was measured during the last 6 h. Strain induced 1.5-2 fold increases in DNA synthesis that were attenuated by antibodies to platelet-derived growth factor (PDGF) AA and BB. Strain also induced increases both in mRNA and protein levels of Sp-1, a transcription factor that binds to the PDGF-A gene promoter site. E2 attenuated strain-induced mitogenesis, and also increases in mRNA and protein levels of Sp-1. The estrogen receptor (ER) antagonist ICI 182,780 (100 nmol/l) reversed the inhibitory effect of E2 on strain-induced increases in DNA synthesis and Sp-1 protein. RT-PCR analysis showed presence of both ER-alpha and -beta in these cells. CONCLUSIONS: Estrogen inhibits strain-induced mitogenesis in human VSM cells via an ER mediated process involving down-regulation of the transcription factor Sp-1.


Asunto(s)
Estradiol/farmacología , Mitosis/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Factor de Transcripción Sp1/metabolismo , Técnicas de Cultivo de Célula , División Celular/fisiología , ADN/biosíntesis , Regulación hacia Abajo , Femenino , Humanos , Músculo Liso Vascular/citología , Músculo Liso Vascular/metabolismo , Factor de Crecimiento Derivado de Plaquetas/genética , Factor de Crecimiento Derivado de Plaquetas/fisiología , ARN Mensajero/genética , Receptores de Estrógenos/genética , Receptores de Estrógenos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Estrés Mecánico
6.
Endocrinology ; 137(10): 4511-3, 1996 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-8828515

RESUMEN

Estrogen is known to modulate angiogenesis, both under physiological and pathological conditions, and has been demonstrated to augment angiogenesis induced by bFGF in a mouse model. We have modified this mouse model and measured the apparent plasma volume in Matrigel plugs containing basic fibroblast growth factor (bFGF) in wild type and estrogen receptor knockout, ovariectomized mice in the presence and absence of exogenous 17 beta estradiol. The apparent plasma volume was determined by measuring the fluorescence of the excised plug 10 min. after injection of fluoroscein labeled dextran 150. In wild type mice exogenous 17 beta estradiol increased the apparent plasma volume of the Matrigel plug and the uterine weight significantly. In the estrogen receptor knockout mice exogenous 17 beta estradiol caused a small, but significant increase in uterine weight but was without effect on the apparent plasma volume of the Matrigel plug. It is concluded that functional estrogen receptors are essential for the augmentation of bFGF-induced angiogenesis by exogenous 17 beta estradiol in female mice.


Asunto(s)
Estradiol/farmacología , Genes , Neovascularización Fisiológica , Receptores de Estrógenos/genética , Animales , Colágeno/química , Combinación de Medicamentos , Femenino , Fluoresceína , Fluoresceínas/análisis , Fluorescencia , Hemoglobinas/análisis , Laminina/química , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Ovariectomía , Proteoglicanos/química , Valores de Referencia , Útero/anatomía & histología
7.
Hypertension ; 25(4 Pt 1): 517-23, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7721392

RESUMEN

We investigated endothelium-dependent responses of thoracic aorta isolated from age-matched male and female spontaneously hypertensive rats (SHR) to explore gender differences in endothelial dysfunction that may contribute to the sexual dimorphism observed in the development of hypertension in this strain. Endothelium-dependent relaxation in response to acetylcholine (10(-9) to 10(-4) mol/L) was significantly greater in female rats than in male rats, although impaired responses were seen in both sexes compared with normotensive controls. Inhibition of cyclooxygenase by indomethacin (10(-5) mol/L) improved endothelium-dependent relaxation, but it did not abolish the gender difference. Relaxations in response to sodium nitroprusside were identical in denuded aortic rings from male and female SHR. Acetylcholine at higher concentrations (10(-6) to 10(-4) mol/L) induced endothelium-dependent contraction in intact, quiescent aortic rings from male SHR but not in those from female SHR. After incubation with NG-nitro-L-arginine (10(-4) mol/L), contraction in response to acetylcholine became apparent in rings from female SHR, but it was still significantly less pronounced than in similarly treated rings from male SHR. Endothelium-dependent contraction was prevented by indomethacin in both sexes, suggesting that a cyclooxygenase product such as endoperoxide may be mediating this effect. Because responses evoked by the thromboxane/endoperoxide receptor agonist U46619 (10(-10) to 10(-6) mol/L) were not greater in rings from male SHR than those from female SHR, increased smooth muscle responsiveness or higher thromboxane/endoperoxide receptor density in the males could not account for the differences in endothelium-dependent contraction. These results suggest that sex steroid hormones may control endothelium-dependent vascular reactivity.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Aorta/fisiopatología , Endotelio Vascular/fisiopatología , Hipertensión/fisiopatología , Caracteres Sexuales , Ácido 15-Hidroxi-11 alfa,9 alfa-(epoximetano)prosta-5,13-dienoico , Acetilcolina/farmacología , Animales , Peso Corporal/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Femenino , Masculino , Óxido Nítrico/biosíntesis , Nitroprusiato/farmacología , Endoperóxidos de Prostaglandinas Sintéticos/farmacología , Ratas , Ratas Endogámicas SHR , Ratas Wistar , Especificidad de la Especie , Tromboxano A2/análogos & derivados , Tromboxano A2/farmacología , Vasoconstrictores/farmacología
8.
Mol Aspects Med ; 22(3): 113-42, 2001 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11470139

RESUMEN

Human gene therapy (HGT) is defined as the transfer of nucleic acids (DNA) to somatic cells of a patient which results in a therapeutic effect, by either correcting genetic defects or by overexpressing proteins that are therapeutically useful. In the past, both the professional and the lay community had high (sometimes unreasonably high) expectations from HGT because of the early promise of treating or preventing diseases effectively and safely by this new technology. Although the theoretical advantages of HGT are undisputable, so far HGT has not delivered the promised results: convincing clinical efficacy could not be demonstrated yet in most of the trials conducted so far, while safety concerns were raised recently as the consequence of the "Gelsinger Case" in Philadelphia. This situation resulted from the by now well-recognized disparity between theory and practice. In other words, the existing technologies could not meet the practical needs of clinically successful HGT so far. However, over the past years, significant progress was made in various enabling technologies, in the molecular understanding of diseases and the manufacturing of vectors. HGT is a complex process, involving multiple steps in the human body (delivery to organs, tissue targeting, cellular trafficking, regulation of gene expression level and duration, biological activity of therapeutic protein, safety of the vector and gene product, to name just a few) most of which are not completely understood. The prerequisite of successful HGT include therapeutically suitable genes (with a proven role in pathophysiology of the disease), appropriate gene delivery systems (e.g., viral and non-viral vectors), proof of principle of efficacy and safety in appropriate preclinical models and suitable manufacturing and analytical processes to provide well-defined HGT products for clinical investigations. The most promising areas for gene therapy today are hemophilias, for monogenic diseases, and cardiovascular diseases (more specifically, therapeutic angiogenesis for myocardial ischemia and peripheral vascular disease, restenosis, stent stenosis and bypass graft failure) among multigenic diseases. This is based on the relative ease of access of blood vessels for HGT, and also because existing gene delivery technologies may be sufficient to achieve effective and safe therapeutic benefits for some of these indications (transient gene expression in some but not all affected cells is required to achieve a therapeutic effect at relatively low [safe] dose of vectors). For other diseases (including cancer) further developments in gene delivery vectors and gene expression systems will be required. It is important to note, that there will not be a "universal vector" and each clinical indication may require a specific set of technical hurdles to overcome. These will include modification of viral vectors (to reduce immunogenicity, change tropism and increase cloning capacity), engineering of non-viral vectors by mimicking the beneficial properties of viruses, cell-based gene delivery technologies, and development of innovative gene expression regulation systems. The technical advances together with the ever increasing knowledge and experience in the field will undoubtedly lead to the realization of the full potential of HGT in the future.


Asunto(s)
Terapia Genética/tendencias , Animales , Predicción , Terapia Genética/efectos adversos , Terapia Genética/legislación & jurisprudencia , Terapia Genética/métodos , Humanos
9.
Free Radic Biol Med ; 4(2): 107-20, 1988.
Artículo en Inglés | MEDLINE | ID: mdl-3278950

RESUMEN

This review attempts to summarize the available data regarding the vascular actions of free oxygen radicals. Studies on blood vessels in situ and in vitro demonstrate that free oxygen radicals can evoke both vasodilation and vasoconstriction. Free oxygen radicals can modulate the tone of vascular smooth muscle by acting directly on the smooth muscle cells, and also via indirect mechanisms by changes in the production or biological activity of vasoactive mediators. The individual oxygen radicals may have different (sometimes opposite) vascular effects. Superoxide anion inactivates endothelium-derived relaxing factor and the adrenergic neurotransmitter norepinephrine. Hydrogen peroxide and the hydroxyl radical evoke vasodilation by acting directly on vascular smooth muscle and also by stimulating the synthesis/release of endothelium-derived relaxing factor. In acute arterial hypertension or experimental brain injury oxygen radicals are important mediators of vascular damage. Production of oxygen-derived free radicals by activated neutrophils may be responsible for vasodilation and increased permeability of capillary membrane during the acute inflammatory process. Free oxygen radicals also play an important role in reperfusion injury of various organs, and vascular actions of the free radicals may contribute to the damage of parenchymal tissues.


Asunto(s)
Vasos Sanguíneos/efectos de los fármacos , Oxígeno/farmacología , Animales , Radicales Libres
10.
J Interferon Cytokine Res ; 18(11): 939-45, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9858315

RESUMEN

The effect of human interferon (IFN)-beta1b (Betaseron) on the proliferation of cultured human vascular smooth muscle and endothelial cells was tested in vitro. IFN-beta1b inhibited thymidine incorporation and growth of primary cultures of human aortic and coronary artery smooth muscle in a concentration-dependent manner. The same concentrations of IFN-beta1b did not inhibit thymidine incorporation or growth of primary cultures of human aortic or coronary artery endothelial cells. IFN-beta1b induced the expression of MxA (an antiviral protein induced by type I IFNs) in both smooth muscle and endothelial cells, suggesting that both cell types express receptors for type I IFNs. The growth-inhibitory effect of IFN-beta1b could be mimicked by commercially available human IFN-beta, but not by IFN-alpha2 or IFN-alpha8. The effect of IFN-beta1b was species specific, as it did not inhibit thymidine incorporation in aortic smooth muscle cells derived from pig, rabbit, rat, or mouse. The action of IFN-beta1b on smooth muscle cells persisted for at least 4 days following a 24 h preincubation with IFN-beta1b. Human vascular smooth muscle cells treated with IFN-beta1b did not release lactate dehydrogenase, nor did they show any morphologic change, suggesting that IFN-beta1b was not toxic to the human vascular smooth muscle cells. IFN-beta1b inhibited vascular smooth muscle growth while having no growth-inhibitory effect on endothelial cells obtained from the same blood vessel, making it a potential candidate for treating pathologic conditions where abnormal vascular smooth muscle proliferation is implicated, such as restenosis following balloon angioplasty or smooth muscle proliferation following vascular stenting.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Endotelio Vascular/efectos de los fármacos , Interferón beta/farmacología , Músculo Liso Vascular/efectos de los fármacos , Animales , Aorta/efectos de los fármacos , División Celular/efectos de los fármacos , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/citología , Humanos , Interferón beta-1a , Interferon beta-1b , Ratones , Músculo Liso Vascular/citología , Conejos , Ratas , Proteínas Recombinantes/farmacología , Porcinos , Timidina/metabolismo
11.
Atherosclerosis ; 144(2): 303-13, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-10407491

RESUMEN

Epidemiological data indicate that estrogens significantly reduce the risk of morbidity and mortality due to cardiovascular diseases in postmenopausal women. Although numerous animal studies demonstrated inhibition of early atheromatous lesion formation by estrogen treatment in several species, information about the potential benefits of estrogens on complex, advanced atherosclerotic lesions is still lacking. The present study was designed to test whether chronic treatment with 17 beta-estradiol affects hyperglycemia-induced premature advanced lesion formation in 40-week-old male apolipoprotein E-deficient (Apo E-KO) mice. In order to accelerate advanced lesion formation, we treated male Apo E-KO mice with streptozotocin (STZ) at the age of 6 weeks. Two weeks later the STZ-treated mice received a slow release pellet containing either 17 beta-estradiol or placebo. STZ treatment caused sustained hyperglycemia without changes in serum total cholesterol or triglyceride levels compared to citrate control mice. STZ-treated Apo E-KO mice developed significantly more lesions in some (but not all) parts of the aorta and its main branches, and caused premature calcified cartilaginous metaplasia in the lesions of the proximal aorta. Chronic treatment with 17 beta-estradiol lead to a significant decrease in blood glucose and triglyceride levels, reduced the lesion area in all vascular segments studied and prevented cartilaginous metaplasia in STZ-treated Apo E-KO mice. The results of this study show that STZ treatment leads to significant acceleration of atherosclerotic lesion formation and premature occurrence of calcified cartilaginous areas in Apo E-KO mice, which could be effectively prevented by chronic estrogen treatment.


Asunto(s)
Apolipoproteínas E/genética , Arteriosclerosis/prevención & control , Calcinosis/prevención & control , Diabetes Mellitus Experimental/prevención & control , Estradiol/farmacología , Animales , Aorta/patología , Arteriosclerosis/genética , Arteriosclerosis/patología , Calcinosis/genética , Calcinosis/patología , Cartílago/patología , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Angiopatías Diabéticas/genética , Angiopatías Diabéticas/patología , Angiopatías Diabéticas/prevención & control , Femenino , Humanos , Masculino , Metaplasia , Ratones , Ratones Noqueados
12.
Br J Pharmacol ; 123(6): 1089-96, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9559891

RESUMEN

1. Studies were performed on isolated aortic rings without endothelium to investigate the effect of 17beta-oestradiol on cytokine-induced nitric oxide production by the inducible nitric oxide synthase (iNOS). 2. Treatment of the isolated aortic rings with interleukin-1beta (IL-1beta, 20 micro ml(-1)) led to the expression of iNOS mRNA and protein, as well as significant nitrite accumulation in the incubation media and suppression of phenylephrine (1 nM-10 microM)-evoked contraction. 3. Cycloheximide (1 microM), a protein synthesis inhibitor, prevented iNOS protein expression, nitrite accumulation and the suppression of contractility by IL-1beta on the isolated aortic rings. 17Beta-oestradiol (1 nM-10 microM) and the partial oestrogen receptor agonist 4-OH-tamoxifen (1 nM-10 microM) produced concentration-dependent inhibition of IL-1beta-induced nitrite accumulation and restored vasoconstrictor responsiveness to phenylephrine, similar to the iNOS inhibitor aminoguanidine (100 microM). 4. Semiquantitative PCR demonstrated decreased iNOS mRNA in the IL-1beta-induced and 17beta-oestradiol-treated rings. Western blot analysis of rat aorta homogenates revealed that 17beta-oestradiol treatment resulted in a reduction in IL-1beta-induced iNOS protein level. 5. Incubation with tumour necrosis factor alpha (TNF alpha, 1 ng ml(-1)) resulted in significant nitrite accumulation in the incubation media and suppression of the smooth muscle contractile response to phenylephrine, similar to IL-1beta. The effects of TNF alpha were also inhibited by co-incubation of the rings with 17beta-oestradiol and 4-OH-tamoxifen (1 microM). 6. The anti-transforming growth factor-beta1 (TGF-beta1) antibody, which inhibited TGF-beta1-induced suppression of nitrite production from IL-1beta-treated vascular rings, did not affect the inhibitory action of 17beta-oestradiol, suggesting that the effect of oestrogen on iNOS inhibition was not mediated by TGF-beta1. 7. These results show that the ovarian sex steroid, 17beta-oestradiol is a modulator of cytokine-induced iNOS activity in rat vascular smooth muscle and its mechanism of action involves decrease of iNOS mRNA and protein.


Asunto(s)
Aorta/efectos de los fármacos , Estradiol/farmacología , Interleucina-1/farmacología , Óxido Nítrico/biosíntesis , Animales , Anticuerpos/inmunología , Aorta/enzimología , Aorta/metabolismo , Técnicas In Vitro , Masculino , Contracción Muscular/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa de Tipo II , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacología , Factor de Crecimiento Transformador beta/inmunología
13.
Shock ; 1(5): 347-53, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-7743337

RESUMEN

The effects of NW-nitro-L-arginine (L-NAG) and dexamethasone in the microcirculatory changes observed in early stages of endotoxemia was investigated in male hamsters treated with Escherichia coli lipopolysaccharide (LPS). The cheek pouch was studied in vivo by means of intravital microscopy and mean arterial and venous pressures, mean arteriolar internal diameter, spontaneous arteriolar vasomotion, microvascular blood flow, macromolecular permeability, leukocyte adhesion, and mean survival time were evaluated in animals treated with either LPS alone or the combination of LPS with L-NAG, an inhibitor of both the constitutive and inducible NO synthases (NOs). The intravenous injection of LPS (100 mg/kg) elicited a significant reduction in mean arterial blood pressure (MABP) and arteriolar blood flow. The observed arterioles dilated and the spontaneous vasomotion ceased. The combination LPS + L-NAG, both given intravenously, prevented the reduction of MABP and the vasodilation but did not help either the reduction of arteriolar blood flow or the cessation of vasomotion. In order to separate the effect of the two NOs, a group of hamsters was pretreated with dexamethasone (10 mg/kg, also intravenously) which inhibits the induction of the inducible NO synthase (iNOs). In this group, the hypotension, vasodilation, and cessation of vasomotion were prevented but the decrease in arteriolar blood flow was not affected. The mean survival time was significantly decreased by the combination of LPS + L-NAG (35 +/- 6 h) and significantly increased by the pretreatment with dexamethasone (92 +/- 5 h) compared to LPS alone (56 +/- 7 h).(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Arginina/análogos & derivados , Permeabilidad Capilar/efectos de los fármacos , Mejilla/irrigación sanguínea , Dexametasona/farmacología , Hemodinámica/efectos de los fármacos , Microcirculación/efectos de los fármacos , Choque Séptico/fisiopatología , Animales , Arginina/farmacología , Adhesión Celular/efectos de los fármacos , Cricetinae , Infecciones por Escherichia coli/fisiopatología , Inyecciones Intravenosas , Lipopolisacáridos/toxicidad , Masculino , Microscopía por Video , Nitroarginina , Choque Séptico/microbiología
14.
Endothelium ; 7(4): 253-64, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-11201523

RESUMEN

Elevated plasma level of lipoprotein(a) (Lp(a)) is a well established risk factor for premature atherosclerosis and coronary artery disease. Recent studies showed impaired endothelium-dependent vasodilatation in humans with elevated plasma Lp(a). However, these human studies could not determine whether (1) elevated Lp(a) levels alone are the cause of endothelial dysfunction (these patients had multiple risk factors), and (2) native or oxidatively modified Lp(a) contributes to endothelial dysfunction (no measurements of native/oxidized Lp(a) ratio was reported in humans). In order to test whether apo(a) (an essential component of Lp(a) which is required for binding to endothelial cells) and native Lp(a) cause endothelial dysfunction, in the present study we tested endothelium-dependent vasorelaxation in aortic rings isolated from control and transgenic male mice either expressing the human apo(a) gene (TgA) or both the human apo(a) and human apo B100 genes (TgL). The TgA mice had plasma apo(a) levels of 8.8 +/- 1.2 mg/dl (n=6) and the double transgenic TgL mice had plasma Lp(a) levels of 15.3 +/- 1.4 mg/dl (n=8). Isolated aortic rings with and without endothelium were mounted in organ chambers and contracted with U46619 (10(-8) M) in the presence of ibuprofen (10(-5) M). Acetylcholine caused concentration-dependent (10(-9)-10(-5) M) relaxation, which could be prevented by endothelium removal and by NG-L-nitro-arginine (10(-4) M). Basal and acetylcholine-stimulated endothelium-dependent relaxation and endothelium-independent relaxation to nitroglycerin (10(-6) M) were not significantly different in aortic rings isolated from control and TgA or TgL mice. Twenty-four hour incubation of aortic rings isolated from control mice with recombinant human apo(a) or native Lp(a) (up to 300 microg/ml) caused no impairment of endothelium-dependent relaxations. In contrast, incubation with oxidized Lp(a) (50 microg/ml) or oxidized LDL (250 microg/ml) caused significant suppression of acetylcholine-induced endothelium-dependent vasorelaxation. These results show for the first time that elevated plasma levels of apo(a) and Lp(a) do not cause endothelial dysfunction in transgenic mice.


Asunto(s)
Aorta/fisiopatología , Apolipoproteínas/fisiología , Endotelio Vascular/fisiopatología , Lipoproteína(a)/fisiología , Vasodilatación , Animales , Aorta/metabolismo , Apolipoproteína B-100 , Apolipoproteínas/genética , Apolipoproteínas B/genética , Apolipoproteínas B/fisiología , Apoproteína(a) , Humanos , Técnicas In Vitro , Lipoproteína(a)/genética , Lipoproteínas LDL/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Óxido Nítrico/metabolismo
15.
Endothelium ; 7(3): 169-84, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10912911

RESUMEN

The goal of this study was to investigate the effect of endothelial cell proliferation on the expression and activity of endothelial nitric oxide synthase (eNOS). Bovine atrial endothelial cells (BAtEC) were studied between day 1 and 6 after seeding. During this period the number of cells in S-phase decreased progressively, while cell number and protein content increased, reaching a maximum at confluence (day 4). Expression of eNOS (determined by ELISA) and eNOS activity (determined by L-arginine to L-citrulline conversion) increased with culture duration with a maximum at confluence. Nitric oxide (*NO) release from BAtEC was determined after stimulation with Ca2+ ionophore A23187 (10 microM, 30 min) by .NO chemiluminescence in the absence of a chemical reduction system. Total *NO release (measured in the presence of 100 U/ml superoxide dismutase) did not change with state of cell proliferation/growth, whereas "bioavailable" *NO (measured in the absence of superoxide dismutase) was low in highly proliferating BAtEC. Relative eNOS activity (.NO and L-citrulline production per eNOS protein) was highest in proliferating BAtEC. The novel finding of this study is that the specific eNOS activity is upregulated in proliferating BAtEC and downregulated in quiescent BAtEC. The amount of "bioavailable" *NO is determined by eNOS activity and *NO inactivation (probably by superoxide), both high in proliferating BAtEC.


Asunto(s)
Endotelio Vascular/citología , Endotelio Vascular/enzimología , Óxido Nítrico Sintasa/metabolismo , Animales , Arginina/metabolismo , Bovinos , División Celular/fisiología , Células Cultivadas , Citrulina/biosíntesis , Endotelio Vascular/efectos de los fármacos , Cinética , Mediciones Luminiscentes , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo III , Nitritos/metabolismo , Superóxido Dismutasa/farmacología , Superóxidos/metabolismo
16.
Neurochem Int ; 18(4): 575-80, 1991.
Artículo en Inglés | MEDLINE | ID: mdl-20504745

RESUMEN

Specific receptor-induced signal transduction mechanisms for the endothelin-2 isoform (ET-2), a potent vasoconstrictor of vascular smooth muscle, were examined in Swiss 3T3 cells. Half-maximal binding (EC(50)) and maximal, saturable binding (B(max)) were estimated from Scatchard analyses and were found to be 24.2 +/- 3.3 pM and 56500 +/- 1700 sites/cells, respectively. A saturating concentration of ET-2 (100 nM) increased intracellular free calcium (measured by Fura-2 fluorescence) from a resting level of ? 100 nM to a peak level of 600-800 nM. The initial increase in intracellular free calcium was transitory and was followed by a smaller maintained elevation (?250 nM). In the absence of extracellular calcium, ET-2 induced a transitory response equal in size to the peak in the presence of extracellular calcium, but the maintained response was absent. ET-2 increased intracellular free calcium in a concentration-dependent manner with an EC(50) of ? 1 nM. In calcium free solution (2 mM EGTA), ET-2 increased the efflux of (45)Ca from cells loaded to isotopic equilibrium (3 h) with (45)Ca. The intracellular second messenger, IP(3), also increased the calcium efflux from saponin permeabilized 3T3 cells loaded with (45)Ca (pCa 6) in the presence of MgATP. In the presence of extracellular calcium, ET-2 significantly increased calcium uptake into 3T3 cells by 92 +/- 36.6 pmoles/million cells/2 min (n = 8). It is suggested that ET-2 binds to specific, high affinity receptors in 3T3 cells and that this receptor interaction increases the intracellular free calcium by IP(3)-induced mobilization of calcium from cellular stores and by increasing influx of extracellular calcium.

17.
Ann N Y Acad Sci ; 522: 226-33, 1988.
Artículo en Inglés | MEDLINE | ID: mdl-3288050

RESUMEN

Indirect and direct experimental evidence demonstrates that both the entry of extracellular calcium and the liberation of calcium from intracellular stores can contribute to an increase in free cytoplasmic calcium concentration in endothelial cells, which seems to be an essential step in the synthesis and/or release of endothelium-derived relaxing factors(s). A variety of Ca2+ transport mechanisms may be involved in the regulation of cytoplasmic calcium in endothelial cells. Ca2+ entry may occur via voltage-operated Ca2+ channels. If they do exist, these channels may have characteristics different from those in underlying vascular smooth muscle cells. Sustained activation of the release of EDRF by various receptor agonists (e.g., acetylcholine, adenine nucleotides, and bradykinin) is also dependent on Ca2+ entry, but it is insensitive to organic Ca2+ channel antagonists. These findings indicate that, when used clinically in various cardiovascular diseases, organic calcium channel antagonists are not expected to interfere with endothelium-dependent relaxation evoked by endogenous vasoactive substances (e.g., ADP, serotonin). Since amiloride and its analogues blocked endothelium-dependent relaxations in different arterial preparations, Na+ transport and Na+/Ca2+ exchange were suggested to play a role in calcium-dependent release of EDRF. The exact nature of Ca2+ transport mechanisms and also the calcium-sensitive cellular processes that lead to the synthesis/release of endothelium derived relaxing factor(s) remain to be determined. However, the available data suggest that calcium handling by the vascular smooth muscle and endothelial cells may be different, allowing potentially selective modulation of Ca2+ activation in these two cell types.


Asunto(s)
Productos Biológicos/metabolismo , Calcio/metabolismo , Animales , Agonistas de los Canales de Calcio/farmacología , Bloqueadores de los Canales de Calcio/farmacología , Endotelio Vascular/metabolismo , Óxido Nítrico , Potasio/farmacología , Conejos , Sodio/metabolismo
18.
Eur J Pharmacol ; 145(3): 351-5, 1988 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-3258243

RESUMEN

Exposure to a K+-free solution reversibly inhibited the acetylcholine-induced relaxation in perfused canine femoral artery segments with endothelium and in superfused bioassay femoral artery rings without endothelium. Infusion of 6.7 mM K+ into the K+-free perfusate downstream of the perfused artery restored the acetylcholine-induced relaxation in the bioassay ring, whereas no relaxation was observed in the perfused segment still exposed to the K+-free solution. These data demonstrate that the K+-free solution depresses the endothelium-dependent relaxation to acetylcholine in femoral arteries by preventing the action, but not the production, of endothelium-derived relaxing factor.


Asunto(s)
Productos Biológicos/biosíntesis , Arteria Femoral/metabolismo , Potasio/metabolismo , Vasodilatadores/biosíntesis , Acetilcolina/farmacología , Animales , Productos Biológicos/farmacología , Dinoprost , Perros , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Arteria Femoral/efectos de los fármacos , Técnicas In Vitro , Masculino , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico , Perfusión , Prostaglandinas F/farmacología , Vasodilatadores/farmacología
19.
Life Sci ; 47(23): 2097-103, 1990.
Artículo en Inglés | MEDLINE | ID: mdl-2266780

RESUMEN

A 21 amino acid peptide containing the prepropendothelin sequence from amino acids 110 to 130 and two intrachain disulfide bonds was synthesized and tested for biological activity in the following endothelin assays: 1.) a competition binding assay using [125I]ET-1 and dog heart membranes, 2.) three RIA's using 125I-ET-1, -2 and -3 and the respective anti-ET rabbit antisera; and 3.) a contractile activity bioassay using hamster aortic rings. The synthetic peptide which has been referred to as the "endothelin-like" peptide occurs 36 amino acids C-terminal to endothelin in the prepro-protein sequence. It contains only 40% sequence homology to the three endothelin isoforms, but has the same sequence and cyclization pattern of cysteines at positions 1, 3, 11 and 15. Despite the overall similarity in secondary structure to the three isoforms of endothelin and sarafotoxin S6b, preproendothelin [110-130] had no activity in any of the assays when tested at concentrations of 10(-10)M to 10(-5)M.


Asunto(s)
Endotelinas/fisiología , Precursores de Proteínas/fisiología , Secuencia de Aminoácidos , Animales , Unión Competitiva , Membrana Celular , Cricetinae , Disulfuros , Perros , Endotelina-1 , Endotelinas/síntesis química , Humanos , Datos de Secuencia Molecular , Miocardio/citología , Precursores de Proteínas/síntesis química , Radioinmunoensayo , Venenos de Víboras/farmacología
20.
J Hypertens Suppl ; 4(5): S290-2, 1986 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-3471908

RESUMEN

Hypertension causes structural and functioning changes in blood vessels. Experiments were performed in isolated hearts of spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats perfused by the Langendorff technique. Spontaneously hypertensive rats had significantly higher left heart to body weight ratios than WKY rats, indicating left ventricular hypertrophy. Coronary flow per unit cardiac mass was lower and vascular resistance was higher at 75 cm H2O perfusion pressure in SHR. This difference was maintained during maximal vasodilatation. In WKY rats, but not in SHR, autoregulation of flow was observed in the pressure range 75-150 cm H2O due to an increase in coronary vascular resistance. After maximal vasodilatation the pressure-flow relationship was linear in SHR and WKY rats, but less steep and shifted to the right in SHR. We conclude that structural adaptations of the coronary circulation in SHR lead to decreased coronary vascular reserve and a loss of autoregulation in the normal blood pressure range.


Asunto(s)
Circulación Coronaria , Corazón/fisiopatología , Homeostasis , Hipertensión/fisiopatología , Animales , Hipertensión/genética , Técnicas In Vitro , Masculino , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA