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











Base de datos
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.
Gene Ther ; 13(18): 1342-50, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16642030

RESUMEN

Gene delivery of angiogenic growth factors is a promising approach for the treatment of ischemic cardiovascular diseases. However, success of this new therapeutic principle is hindered by the lack of critical understanding as to how disease pathology affects the efficiency of gene delivery and/or the downstream signaling pathways of angiogenesis. Critical limb ischemia occurs in patients with advanced atherosclerosis often exhibiting deficiency in endothelial nitric oxide production. Similar to these patients, segmental femoral artery resection progresses into severe ischemic necrosis in mice deficient in endothelial nitric oxide synthase (ecNOS-KO) as well as in balb/c mice. We used these models to evaluate the influence of severe ischemia on transfection efficiency and duration of transgene expression in the skeletal muscle following plasmid injection in combination with electroporation. Subsequently, we also explored the potential therapeutic effect of the phosphomimetic mutant of ecNOS gene (NOS1177D) using optimized delivery parameters, and found significant benefit both in ecNOS-KO and balb/c mice. Our results indicate that NOS1177D gene delivery to the ischemic skeletal muscle can be efficient to reverse critical limb ischemia in pathological settings, which are refractory to treatments with a single growth factor, such as vascular endothelial growth factor.


Asunto(s)
Terapia Genética/métodos , Isquemia/terapia , Músculo Esquelético/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Transfección/métodos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Electroporación , Endotelio Vascular/metabolismo , Expresión Génica , Vectores Genéticos , Miembro Posterior , Humanos , Isquemia/metabolismo , Isquemia/patología , Flujometría por Láser-Doppler , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/patología , Neovascularización Fisiológica , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/análisis , Óxido Nítrico Sintasa de Tipo III/metabolismo , Flujo Sanguíneo Regional , Transgenes , Vasodilatación
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
5.
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
6.
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
7.
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
9.
Am J Physiol Heart Circ Physiol ; 278(5): H1679-85, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10775149

RESUMEN

This study investigated the role of endogenous nitric oxide (NO) in the progression of atherosclerosis in apolipoprotein E-deficient [apoE-knockout (KO)] mice. Mice were treated with N(omega)-nitro-L-arginine methyl ester (L-NAME) an inhibitor of nitric oxide synthase (NOS) or with the NOS substrate L-arginine for 8 wk. L-NAME treatment resulted in a significant inhibition of NO-mediated vascular responses and a significant increase in the atherosclerotic plaque/surface area in the aorta of apoE-KO mice. L-arginine treatment had no influence on endothelial function and did not alter lesion size. Mean arterial blood pressure and serum lipid levels were not altered by the treatments. At the beginning of the study impairment in endothelial function was only apparent in the case of N(G)-nitro-L-arginine-induced, NO-mediated contraction, whereas ACh-induced, NO-mediated relaxation was not different between age-matched apoE-KO and C57Bl/6J mice. After the 8-wk treatment with the NOS inhibitor, both NO-mediated responses were significantly inhibited. The acceleration in lesion size concomitant to the severely impaired NO-mediated responses indicates that lack of endogenous NO is an important progression factor of atherosclerosis in the apoE-KO mouse.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/metabolismo , Óxido Nítrico/metabolismo , Acetilcolina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/metabolismo , Aorta/patología , Apolipoproteínas E/genética , Arginina/farmacología , Arteriosclerosis/genética , Arteriosclerosis/patología , Presión Sanguínea/efectos de los fármacos , Progresión de la Enfermedad , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Inhibidores Enzimáticos/farmacología , Técnicas In Vitro , Lípidos/sangre , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , NG-Nitroarginina Metil Éster/farmacología , Nitroarginina/farmacología , Nitroprusiato/farmacología , Vasoconstricción/efectos de los fármacos
10.
J Biol Chem ; 275(21): 15885-94, 2000 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-10748166

RESUMEN

Clinical studies have shown that estrogen replacement therapy (ERT) reduces the incidence and severity of osteoporosis and cardiovascular disease in postmenopausal women. However, long term estrogen treatment also increases the risk of endometrial and breast cancer. The selective estrogen receptor (ER) modulators (SERMs) tamoxifen and raloxifene, cause antagonistic and agonistic responses when bound to the ER. Their predominantly antagonistic actions in the mammary gland form the rationale for their therapeutic utility in estrogen-responsive breast cancer, while their agonistic estrogen-like effects in bone and the cardiovascular system make them candidates for ERT regimens. Of these two SERMs, raloxifene is preferred because it has markedly less uterine-stimulatory activity than either estrogen or tamoxifen. To identify additional SERMs, a method to classify compounds based on differential gene expression modulation was developed. By analysis of 24 different combinations of genes and cells, a selected set of assays that permitted discrimination between estrogen, tamoxifen, raloxifene, and the pure ER antagonist ICI164384 was generated. This assay panel was employed to measure the activity of 38 compounds, and the gene expression fingerprints (GEFs) obtained for each compound were used to classify all compounds into eight groups. The compound's GEF predicted its uterine-stimulatory activity. One group of compounds was evaluated for activity in attenuating bone loss in ovariectomized rats. Most compounds with similar GEFs had similar in vivo activities, thereby suggesting that GEF-based screens could be useful in predicting a compound's in vivo pharmacological profile.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Receptores de Estrógenos/metabolismo , Moduladores Selectivos de los Receptores de Estrógeno/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Endometrio/efectos de los fármacos , Estradiol/análogos & derivados , Estradiol/farmacología , Terapia de Reemplazo de Estrógeno , Estrógenos/farmacología , Femenino , Humanos , Alcamidas Poliinsaturadas , Unión Proteica , ARN Mensajero/metabolismo , Clorhidrato de Raloxifeno/farmacología , Clorhidrato de Raloxifeno/uso terapéutico , Ratas , Ratas Wistar , Tamoxifeno/farmacología , Tamoxifeno/uso terapéutico , Transcortina/genética , Células Tumorales Cultivadas
11.
Am J Physiol Heart Circ Physiol ; 278(2): H428-34, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10666072

RESUMEN

Atherosclerosis develops and progresses spontaneously in apolipoprotein E-knockout (apoE-KO) mice. A direct consequence of atherosclerosis is an increase in vascular stiffness. Pulse wave velocity (PWV) has been used to assess the stiffness of large vessels and was found to be increased in patients with atherosclerosis. In the present study, aortic stiffness was assessed by PWV in 4- and 13-mo-old apoE-KO mice and age-matched controls (C57BL/6J). In 13-mo-old apoE-KO mice with extensive atherosclerotic lesions in the aorta (61 +/- 4%), PWV increased significantly (3.8 +/- 0.2 m/s) compared with controls (2.9 +/- 0.2 m/s). Endothelial nitric oxide (EDNO)-mediated vasorelaxation in response to ACh was markedly diminished in the aortic rings isolated from 13-mo-old apoE-KO mice compared with age-matched controls. In contrast, in 4-mo-old apoE-KO mice with only moderate atherosclerotic lesions in the aorta (23 +/- 5%), there were no significant changes in PWV and EDNO-mediated relaxation compared with controls. Blood pressure was not different among the four groups of mice. There were no significant differences in endothelium-independent vascular responses to sodium nitroprusside among different groups investigated. Histological evaluation revealed focal fragmentation of the elastic laminae in the aortic walls of 13-mo-old apoE-KO mice. These results demonstrate for the first time that aortic stiffness determined by PWV increases in 13-mo-old apoE-KO mice. Endothelial dysfunction and elastic destruction in vascular wall caused by atherosclerosis may have contributed.


Asunto(s)
Aorta/fisiopatología , Apolipoproteínas E/deficiencia , Pulso Arterial , Acetilcolina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/patología , Enfermedades de la Aorta/fisiopatología , Apolipoproteínas E/genética , Arteriosclerosis/fisiopatología , Elasticidad , Endotelio Vascular/fisiopatología , Femenino , Técnicas In Vitro , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados/genética , Vasodilatación , Vasodilatadores/farmacología
12.
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
13.
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
14.
Am J Physiol ; 276(5): H1788-94, 1999 05.
Artículo en Inglés | MEDLINE | ID: mdl-10330264

RESUMEN

17beta-Estradiol prevents early vascular lesion development and may also affect advanced atherosclerosis. To test the antiatherosclerotic effect of estrogen under conditions that resemble more advanced human atherosclerosis with severe endothelial dysfunction, we have investigated the effect of 17beta-estradiol in hypercholesterolemic rabbits treated with the nitric oxide synthase inhibitor Nomega-nitro-L-arginine methyl ester (L-NAME). Chronic L-NAME administration attenuated endothelial nitric oxide (EDNO)-mediated vascular responses leading to significantly accelerated atherosclerotic plaque development. 17beta-Estradiol treatment alone inhibited aortic lesion formation with concurrent increase in EDNO-mediated responses. The beneficial effect of estrogen persisted in the L-NAME-treated rabbits, suggesting that the antiatherogenic action of 17beta-estradiol involves NO-independent mechanisms as well. Serum cholesterol levels were not altered by any of the treatments. 17beta-Estradiol treatment significantly increased EDNO production under these conditions as well. The reduction in plaque size by 17beta-estradiol was always accompanied by increased EDNO production, suggesting a strong association between these two events. The results demonstrate that estrogen treatment may exert protection against atherosclerosis even in patients with severe endothelial dysfunction.


Asunto(s)
Endotelio Vascular/fisiopatología , Estradiol/farmacología , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/metabolismo , Acetilcolina/farmacología , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Arteriosclerosis/tratamiento farmacológico , Arteriosclerosis/metabolismo , Colesterol/sangre , Endotelio Vascular/enzimología , Inhibidores Enzimáticos/farmacología , Estradiol/sangre , Femenino , Técnicas In Vitro , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Nitroglicerina/farmacología , Conejos , Vasodilatación/efectos de los fármacos , Vasodilatación/fisiología , Vasodilatadores/farmacología
15.
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
16.
Arterioscler Thromb Vasc Biol ; 18(9): 1498-505, 1998 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9743240

RESUMEN

Increased levels of interleukin-6 (IL-6) have been proposed to contribute to a number of pathological disorders, including osteoporosis and Alzheimer's disease. In human atherosclerotic lesions, IL-6 protein and mRNA have been detected, although the role of IL-6 in plaque formation is unknown. We have examined the expression pattern of IL-6 mRNA and secreted protein in male apolipoprotein E-knockout (apoE-KO) mice aortas. Furthermore, we have evaluated the effects of 17beta-estradiol (E2), a vasculoprotective sex steroid hormone, on the secretion of this inflammatory cytokine from isolated male apoE-KO mice aortas. The expression of IL-6 mRNA was detected by reverse transcription-polymerase chain reaction in the apoE-KO mouse aortas but not in the aortas of age-matched control mice. Similarly, the secretion of IL-6 protein from isolated apoE-KO aortic segments was significantly greater than that from aortas of age-matched control animals. The secretion of IL-6 from isolated aortic rings of apoE-KO mice ranging in age from 6 to 48 weeks showed a significant, positive correlation with percent lesion area measured in the same tissue. Immunohistochemical staining of apoE-KO mouse aortic tissue sections demonstrated colocalization of IL-6 expression with macrophages. Treatment of male apoE-KO mice with E2 for 3 weeks resulted in a statistically significant 50% reduction in IL-6 secretion from ex vivo aortic tissue segments. There was no significant change in total serum cholesterol and triglyceride levels in the E2-treated group compared with placebo-treated controls. These data demonstrate that (1) IL-6 mRNA and protein are expressed in the atherosclerotic plaques of apoE-KO mice aortas and (2) IL-6 production is suppressed by E2 treatment, which may contribute to the antiatherosclerotic effects of E2 in the apoE-KO mouse model of atherosclerosis.


Asunto(s)
Apolipoproteínas E/deficiencia , Arteriosclerosis/metabolismo , Estradiol/farmacología , Expresión Génica/efectos de los fármacos , Interleucina-6/genética , Animales , Aorta/química , Aorta/efectos de los fármacos , Aorta/metabolismo , Apolipoproteínas E/genética , Colesterol/sangre , Interleucina-6/análisis , Interleucina-6/metabolismo , Macrófagos/química , Masculino , Ratones , Ratones Noqueados , Técnicas de Cultivo de Órganos , Reacción en Cadena de la Polimerasa , ARN Mensajero/análisis , ADN Polimerasa Dirigida por ARN , Triglicéridos/sangre
17.
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
19.
Circulation ; 96(10): 3774-7, 1997 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-9396482

RESUMEN

BACKGROUND: While estrogens protect against coronary artery disease in women, it is unclear whether they influence cardiovascular function in men. The present report describes coronary vascular abnormalities and the lipoprotein profile of a male patient with estrogen insensitivity caused by a disruptive mutation in the estrogen-receptor gene. METHODS AND RESULTS: Stress thallium scintigraphy, echocardiography, and electron-beam computed tomography (CT) scanning of the coronary arteries and detailed lipoprotein analysis were performed. Electron-beam CT scanning of the coronary arteries showed calcium in the left anterior descending artery. Lipoprotein analysis showed relatively low levels of total (130 mg/dL), LDL (97 mg/dL), and HDL (34 mg/dL) cholesterol; apolipoprotein A-I (91.7 mg/dL); and lipoprotein(a) (4.1 nmol/L), but normal levels of triglycerides (97 mg/dL) and pre-beta-1-HDL cholesterol (61 microg/mL). CONCLUSIONS: The absence of functional estrogen receptors may be a novel risk factor for coronary artery disease in men.


Asunto(s)
Enfermedad Coronaria/genética , Mutación/fisiología , Receptores de Estrógenos/genética , Adulto , Enfermedad Coronaria/diagnóstico , Ecocardiografía , Prueba de Esfuerzo , Humanos , Lipoproteínas/sangre , Masculino , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X
20.
Arterioscler Thromb Vasc Biol ; 17(9): 1741-5, 1997 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-9327772

RESUMEN

Plasma levels of lipoprotein(a) (Lp(a)), are regulated by the synthetic rate of apolipoprotein(a) (apo(a)), a major protein component of this atherogenic lipoprotein. Exogenously administered sex steroid hormones are potent regulators of plasma Lp(a) concentrations. We utilized a recently developed apo(a) yeast artificial chromosome (YAC) transgenic mouse model to study the effects of ovariectomy, estrus cycle, and exogenous administration of ethinyl-estradiol, the partial estrogen receptor agonist, tamoxifen, and progesterone on circulating apo(a) plasma levels. Analysis of liver RNA revealed that estrogen and tamoxifen exerts their plasma apo(a) lowering effect at the level of apo(a) mRNA. This action of estrogen and tamoxifen may contribute to their antiatherosclerotic and cardiovascular protective effect.


Asunto(s)
Apolipoproteínas/genética , Congéneres del Estradiol/farmacología , Antagonistas de Estrógenos/farmacología , Estro/fisiología , Etinilestradiol/farmacología , Expresión Génica/efectos de los fármacos , Lipoproteína(a) , Progesterona/farmacología , Tamoxifeno/farmacología , Animales , Apolipoproteínas/sangre , Apoproteína(a) , Combinación de Medicamentos , Femenino , Expresión Génica/fisiología , Hígado/metabolismo , Ratones , Ratones Transgénicos/genética , Ovariectomía , ARN Mensajero/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA