Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 310(10): H1285-94, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-26993224

RESUMEN

In the present study, interactions of age and estrogen in the modulation of cerebrovascular function were examined in small arteries <150 µM. The hypothesis tested was that age enhances deleterious effects of exogenous estrogen by augmenting constrictor prostanoid (CP)-potentiated reactivity of the female (F) cerebrovasculature. F Sprague-Dawley rats approximating key stages of "hormonal aging" in humans were studied: perimenopausal (mature multi-gravid, MA, cyclic, 5-6 mo of age) and postmenopausal (reproductively senescent, RS, acyclic 10-12 mo of age). Rats underwent bilateral ovariectomy and were given estrogen replacement therapy (E) or placebo (O) for 14-21 days. Vasopressin reactivity (VP, 10(-12)-10(-7) M) was measured in pressurized middle cerebral artery segments, alone or in the presence of COX-1- (SC560, 1 µM) or COX-2- (NS398, 10 µM) selective inhibitors. VP-stimulated release of prostacyclin (PGI2) and thromboxane (TXA2) were assessed by radioimmunoassay of 6-keto-PGF1α and TXB2 (stable metabolites). VP-induced vasoconstriction was attenuated in ovariectomized + estrogen-replaced, multigravid adult rats (5-6 mo; MAE) but potentiated in older ovariectomized + estrogen-replaced, reproductively senescent rats (12-14 mo; RSE). SC560 and NS398 reduced reactivity similarly in ovariectomized multigravid adult rats (5-6 mo; MAO) and ovariectomized reproductively senescent rat (12-14 mo; RSO). In MAE, reactivity to VP was reduced to a greater extent by SC560 than by NS398; however, in RSE, this effect was reversed. VP-stimulated PGI2 was increased by estrogen, yet reduced by age. VP-stimulated TXA2 was increased by estrogen and age in RSE but did not differ in MAO and RSO. Taken together, these data reveal that the vascular effects of estrogen are distinctly age-dependent in F rats. In younger MA, beneficial and protective effects of estrogen are evident (decreased vasoconstriction, increased dilator prostanoid function). Conversely, in older RS, detrimental effects of estrogen begin to be manifested (enhanced vasoconstriction and CP function). These findings may lead to age-specific estrogen replacement therapies that maximize beneficial and minimize detrimental effects of this hormone on small cerebral arteries that regulate blood flow.


Asunto(s)
Estradiol/administración & dosificación , Terapia de Reemplazo de Estrógeno , Arteria Cerebral Media/efectos de los fármacos , Vasoconstricción/efectos de los fármacos , Factores de Edad , Animales , Relación Dosis-Respuesta a Droga , Epoprostenol/metabolismo , Estradiol/toxicidad , Terapia de Reemplazo de Estrógeno/efectos adversos , Arteria Cerebral Media/metabolismo , Ovariectomía , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tromboxano A2/metabolismo , Vasoconstrictores/farmacología , Vasopresinas/farmacología
2.
Am J Physiol Regul Integr Comp Physiol ; 309(2): R189-95, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-25947172

RESUMEN

Testosterone (TES) and other androgens exert a direct vasorelaxing action on the vasculature in vitro that is structurally specific and independent of cytosolic androgen receptor (AR). The effects of intravenous androgen infusions on mean arterial blood pressure (BP) and heart rate (HR) were determined in conscious, unrestrained, chronically catheterized, ganglionically blocked (hexamethonium, HEX; 30 mg/kg ip) male Sprague-Dawley (SD) and testicular-feminized male (Tfm; AR-deficient) rats, 16-20 wk of age. BP and HR were recorded at baseline and with increasing doses of androgens (0.375-6.00 µmol·kg(-1)·min(-1) iv; 10 min/dose). Data are expressed as means ± SE (n = 5-8 rats/group). In SD rats, baseline BP and HR averaged 103 ± 4 mmHg and 353 ± 12 beats/min (bpm). TES produced a dose-dependent reduction in BP to a low of 87 ± 4 mmHg (Δ16%), while HR was unchanged (354 ± 14 bpm). Neither BP (109 ± 3 mmHg) nor HR (395 ± 13 bpm) were altered by vehicle (10% EtOH in 0.9% saline; 0.15 ml·kg(-1)·min(-1), iv). In Tfm, TES produced a similar reduction in BP (99 ± 3 to 86 ± 3 mmHg, Δ13%); HR was unchanged (369 ± 18 bpm). In SD, 5ß-dihydrotestosterone (genomically inactive metabolite) produced a greater reduction in BP than TES (102 ± 2 to 79 ± 2 mmHg, Δ23%); HR was unchanged (361 ± 9). A 20-µg iv bolus of sodium nitroprusside in both SD and Tfm rats reduced BP 30-40 mmHg, while HR was unchanged, confirming blockade by HEX. Pretreatment of SD rats with neuronal nitric oxide synthase (nNOS) inhibitor (S-methyl-thiocitrulline, SMTC; 20 µg·kg(-1)·min(-1) × 30 min) abolished the hypotensive effects of TES infusion on BP (104 ± 2 vs. 101 ± 2 mmHg) and HR (326 ± 11 vs. 324 ± 8 bpm). These data suggest the systemic hypotensive effect of TES and other androgens involves a direct vasodilatory action on the peripheral vasculature which, like the effect observed in isolated arteries, is structurally specific and AR-independent, and involves activation of nNOS.


Asunto(s)
Andrógenos/administración & dosificación , Presión Arterial/efectos de los fármacos , Arterias/efectos de los fármacos , Hipotensión/inducido químicamente , Óxido Nítrico Sintasa de Tipo I/metabolismo , Testosterona/administración & dosificación , Síndrome de Resistencia Androgénica/genética , Síndrome de Resistencia Androgénica/metabolismo , Síndrome de Resistencia Androgénica/fisiopatología , Andrógenos/química , Animales , Arterias/enzimología , Arterias/fisiopatología , Dihidrotestosterona/administración & dosificación , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Frecuencia Cardíaca/efectos de los fármacos , Hipotensión/enzimología , Hipotensión/fisiopatología , Infusiones Intravenosas , Masculino , Estructura Molecular , Óxido Nítrico Sintasa de Tipo I/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Relación Estructura-Actividad , Testosterona/análogos & derivados , Testosterona/química , Factores de Tiempo , Vasodilatación/efectos de los fármacos
3.
Am J Physiol Endocrinol Metab ; 307(4): E398-407, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25005496

RESUMEN

Activation of GPER exerts a protective effect in hypertension and ischemia-reperfusion models and relaxes arteries in vitro. However, our understanding of the mechanisms of GPER-mediated vascular regulation is far from complete. In the current study, we tested the hypothesis that GPER-induced relaxation of porcine coronary arteries is mediated via cAMP/PKA signaling. Our findings revealed that vascular relaxation to the selective GPER agonist G-1 (0.3-3 µM) was associated with increased cAMP production in a concentration-dependent manner. Furthermore, inhibition of adenylyl cyclase (AC) with SQ-22536 (100 µM) or of PKA activity with either Rp-8-CPT-cAMPS (5 µM) or PKI (5 µM) attenuated G-1-induced relaxation of coronary arteries preconstricted with PGF2α (1 µM). G-1 also increased PKA activity in cultured coronary artery smooth muscle cells (SMCs). To determine downstream signals of the cAMP/PKA cascade, we measured RhoA activity in cultured human and porcine coronary SMCs and myosin-light chain phosphatase (MLCP) activity in these artery rings by immunoblot analysis of phosphorylation of myosin-targeting subunit protein-1 (p-MYPT-1; the MLCP regulatory subunit). G-1 decreased PGF2α-induced p-MYPT-1, whereas Rp-8-CPT-cAMPS prevented this inhibitory effect of G-1. Similarly, G-1 inhibited PGF2α-induced phosphorylation of MLC in coronary SMCs, and this inhibitory effect was also reversed by Rp-8-CPT-cAMPS. RhoA activity was downregulated by G-1, whereas G36 (GPER antagonist) restored RhoA activity. Finally, FMP-API-1 (100 µM), an inhibitor of the interaction between PKA and A-kinase anchoring proteins (AKAPs), attenuated the effect of G-1 on coronary artery relaxation and p-MYPT-1. These findings demonstrate that localized cAMP/PKA signaling is involved in GPER-mediated coronary vasodilation by activating MLCP via inhibition of RhoA pathway.


Asunto(s)
Vasos Coronarios/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Receptores de Estrógenos/fisiología , Receptores Acoplados a Proteínas G/fisiología , Vasodilatación , Animales , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , AMP Cíclico/metabolismo , AMP Cíclico/farmacología , Activación Enzimática/efectos de los fármacos , Humanos , Transducción de Señal/efectos de los fármacos , Porcinos , Vasodilatación/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 345(1): 7-14, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23318471

RESUMEN

Our knowledge of how androgens influence the cardiovascular system is far from complete, and this lack of understanding is especially true of how androgens affect resistance vessels. Our aim was to identify the signaling mechanisms stimulated by testosterone (TES) in microvascular arteries and to understand how these mechanisms mediate TES-induced vasodilation. Mesenteric microvessels were isolated from male Sprague-Dawley rats. Tension studies demonstrated a rapid, concentration-dependent, vasodilatory response to TES that did not involve protein synthesis or aromatization to 17ß-estradiol. Dichlorofluorescein fluorescence and nitrotyrosine immunoblot experiments indicated that TES stimulated peroxynitrite formation in microvessels, and functional studies demonstrated that TES-induced vasodilation was inhibited by scavenging peroxynitrite. As predicted, TES enhanced the production of both peroxynitrite precursors (i.e., superoxide and nitic oxide), and xanthine oxidase was identified as the likely source of TES-stimulated superoxide production. Functional and biochemical studies indicated that TES signaling involved activity of the phosphoinositide 3 (PI3) kinase-protein kinase B (Akt) cascade initiated by activation of the androgen receptor and culminated in enhanced production of cGMP and microvascular vasodilation. These findings, derived from a variety of analytical and functional approaches, provide evidence for a novel nongenomic signaling mechanism for androgen action in the microvasculature: TES-stimulated vasodilation mediated primarily by peroxynitrite formed from xanthine oxidase-generated superoxide and NO. This response was associated with activation of the PI3 kinase-Akt signaling cascade initiated by activation of the androgen receptor. We propose this mechanism could account for TES-stimulated cGMP production in microvessels and, ultimately, vasodilation.


Asunto(s)
Andrógenos/farmacología , Microvasos/efectos de los fármacos , Ácido Peroxinitroso/biosíntesis , Testosterona/farmacología , Resistencia Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Animales , Western Blotting , GMP Cíclico/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Técnicas In Vitro , Masculino , Microvasos/metabolismo , Microvasos/fisiopatología , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Superóxidos/metabolismo , Xantina Oxidasa/metabolismo
5.
Biochem Pharmacol ; 208: 115347, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36395900

RESUMEN

Cardiovascular disease (CVD) is a major cause of morbidity and mortality worldwide and in the Western world, one-third of all deaths are attributed to CVD. A conspicuous characteristic of this healthcare epidemic is that most CVD is higher in men than in age-matched premenopausal women, yet reasons for these obvious sex differences remain poorly understood. Driven by clinical case and epidemiological studies and supported by animal experiments, a strong dogma emerged early on that testosterone (TES) exerts deleterious effects on cardiovascular health and exacerbates development of CVD and metabolic dysfunctions in men. In this review, earlier and more recent clinical and experimental animal evidence of cardiovascular and metabolic effects of androgens are discussed. The more recent evidence overwhelmingly suggests that it is progressive, age-dependent declines in TES levels in men that exacerbate CVD and metabolic dysfunctions, while TES exerts beneficial systemic hypotensive effects and protects against metabolic syndrome (MetS) and type2 diabetes mellitus (T2DM). Recent findings reveal existence of bi-directional modulation of glucose and fat homeostasis by TES in females vs males, such that age-dependent declines in TES levels in males and abnormal increases in normally low TES levels in females both result in similar dysfunction in glucose and fat homeostasis, resulting in development of MetS and T2DM, central risk factors for development of CVD, in men as well as women. These findings suggest that the long-held view that TES is detrimental to male health should be discarded in favor of the view that, at least in men, TES is beneficial to cardiovascular and metabolic health.


Asunto(s)
Enfermedades Cardiovasculares , Diabetes Mellitus Tipo 2 , Síndrome Metabólico , Animales , Femenino , Masculino , Andrógenos/metabolismo , Testosterona/metabolismo , Glucosa
6.
Am J Physiol Heart Circ Physiol ; 302(1): H115-23, 2012 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-22081702

RESUMEN

Androgens are reported to have both beneficial and detrimental effects on human cardiovascular health. The aim of this study was to characterize nongenomic signaling mechanisms in coronary artery smooth muscle (CASM) and define the ionic basis of testosterone (TES) action. TES-induced relaxation of endothelium-denuded porcine coronary arteries was nearly abolished by 20 nM iberiotoxin, a highly specific inhibitor of large-conductance, calcium-activated potassium (BK(Ca)) channels. Molecular patch-clamp studies confirmed that nanomolar concentrations of TES stimulated BK(Ca) channel activity by ∼100-fold and that inhibition of nitric oxide synthase (NOS) activity by N(G)-monomethyl-L-arginine nearly abolished this effect. Inhibition of nitric oxide (NO) synthesis or guanylyl cyclase activity also attenuated TES-induced coronary artery relaxation but did not alter relaxation due to 8-bromo-cGMP. Furthermore, we detected TES-stimulated NO production in porcine coronary arteries and in human CASM cells via stimulation of the type 1 neuronal NOS isoform. Inhibition of the cGMP-dependent protein kinase (PKG) attenuated TES-stimulated BK(Ca) channel activity, and direct assay determined that TES increased activity of PKG in a concentration-dependent fashion. Last, the stimulatory effect of TES on BK(Ca) channel activity was mimicked by addition of purified PKG to the cytoplasmic surface of a cell-free membrane patch from CASM myocytes (∼100-fold increase). These findings indicate that TES-induced relaxation of endothelium-denuded coronary arteries is mediated, at least in part, by enhanced NO production, leading to cGMP synthesis and PKG activation, which, in turn, opens BK(Ca) channels. These findings provide a molecular mechanism that could help explain why androgens have been reported to relax coronary arteries and relieve angina pectoris.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Músculo Liso Vascular/enzimología , Testosterona/metabolismo , Vasodilatación , Animales , Vasos Coronarios/enzimología , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Relación Dosis-Respuesta a Droga , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Femenino , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/antagonistas & inhibidores , Masculino , Potenciales de la Membrana , Músculo Liso Vascular/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Técnicas de Placa-Clamp , Fosforilación , Canales de Potasio/farmacología , Transducción de Señal , Porcinos , Factores de Tiempo , Vasodilatación/efectos de los fármacos
7.
Steroids ; 183: 108997, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35314416

RESUMEN

BACKGROUND: Hormone replacement therapy was found to be effective in cardiovascular protection only in younger women, not in older women. In this study, we tested whether G protein-coupled estrogen receptor 1 (GPER) activation improves vascular activities in response to ET-1 and ACh in aging rats. METHODS: Isometric tension study was applied on aortic rings isolated from young adult (5-7 months) and reproductive senescent middle-aged (10-12 months) female Sprague Dawley rats and age matched males. RESULTS: The aortic contractile response to ET-1 and the relaxation response to ACh were reduced in the female middle-aged rats compared to the female young adult rats. The presence of G-1, the GPER agonist, normalized the reduced vascular activities. Cyclooxygenase inhibitor, meclofenamate, blocked the increased constriction effect of G-1, but further enhanced relaxation effect of G-1. There was no significant difference in aortic reactivity to either ET-1 or ACh between the male middle-aged and young adult rats. The contractile response to ET-1 was not different within the same age of the two sex groups, but there was a remarkable difference in relaxation response to ACh between young adult females and males with better response in females. GPER activation greatly improved the aortic relaxation of both young adult and middle-aged females, but not the males. CONCLUSIONS: Endothelial dysfunction occurs earlier in males, but in females, dysfunction delays until middle age. GPER activation improves the vascular activities in females, but not males. It is promising to employ GPER as a potential drug target in cardiovascular disease in women.


Asunto(s)
Receptores de Estrógenos , Receptores Acoplados a Proteínas G , Anciano , Animales , Endotelio Vascular , Estrógenos/farmacología , Femenino , Proteínas de Unión al GTP/farmacología , Humanos , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
8.
Physiol Genomics ; 43(18): 1065-73, 2011 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-21750230

RESUMEN

Despite an abundance of evidence to the contrary from animal studies, large clinical trials on humans have shown that estrogen administered to postmenopausal women increases the risk of cardiovascular disease. However, timing may be everything, as estrogen is often administered immediately after ovariectomy (Ovx) in animal studies, while estrogen administration in human studies occurred many years postmenopause. This study investigates the discrepancy by administering 17ß-estradiol (E2) in a slow-release capsule to Norway Brown rats both immediately following Ovx and 9 wk post-Ovx (Late), and studying differences in gene expression between these two groups compared with age-matched Ovx and sham-operated animals. Two different types of microarray were used to analyze the left ventricles from these groups: an Affymetrix array (n = 3/group) and an inflammatory cytokines and receptors PCR array (n = 4/group). Key genes were analyzed by Western blotting. Ovx without replacement led to an increase in caspase 3, caspase 9, calpain 2, matrix metalloproteinase (MMP)9, and TNF-α. Caspase 6, STAT3, and CD11b increased in the Late group, while tissue inhibitor of metalloproteinase 2, MMP14, and collagen I α1 were decreased. MADD and fibronectin were increased in both Ovx and Late. TNF-α and inducible nitric oxide synthase (iNOS) protein levels increased with Late replacement. Many of these changes were prevented by early E2 replacement. These findings suggest that increased expression of inflammatory genes, such as TNF-α and iNOS, may be involved in some of the deleterious effects of delayed E2 administration seen in human studies.


Asunto(s)
Envejecimiento/sangre , Terapia de Reemplazo de Estrógeno , Estrógenos/sangre , Estrógenos/uso terapéutico , Regulación de la Expresión Génica , Inflamación/tratamiento farmacológico , Inflamación/genética , Miocardio/metabolismo , Animales , Apoptosis/genética , Western Blotting , Matriz Extracelular/genética , Femenino , Humanos , Modelos Biológicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Ratas , Ratas Endogámicas BN , Transducción de Señal/genética
9.
Am J Physiol Endocrinol Metab ; 301(5): E882-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21791623

RESUMEN

Estrogens can either relax or contract arteries via rapid, nongenomic mechanisms involving classic estrogen receptors (ER). In addition to ERα and ERß, estrogen may also stimulate G protein-coupled estrogen receptor 1 (GPER) in nonvascular tissue; however, a potential role for GPER in coronary arteries is unclear. The purpose of this study was to determine how GPER activity influenced coronary artery reactivity. In vitro isometric force recordings were performed on endothelium-denuded porcine arteries. These studies were augmented by RT-PCR and single-cell patch-clamp experiments. RT-PCR and immunoblot studies confirmed expression of GPER mRNA and protein, respectively, in smooth muscle from either porcine or human coronary arteries. G-1, a selective GPER agonist, produced a concentration-dependent relaxation of endothelium-denuded porcine coronary arteries in vitro. This response was attenuated by G15, a GPER-selective antagonist, or by inhibiting large-conductance calcium-activated potassium (BK(Ca)) channels with iberiotoxin, but not by inhibiting NO signaling. Last, single-channel patch-clamp studies demonstrated that G-1 stimulates BK(Ca) channel activity in intact smooth muscle cells from either porcine or human coronary arteries but had no effect on channels isolated in excised membrane patches. In summary, GPER activation relaxes coronary artery smooth muscle by increasing potassium efflux via BK(Ca) channels and requires an intact cellular signaling mechanism. This novel action of estrogen-like compounds may help clarify some of the controversy surrounding the vascular effects of estrogens.


Asunto(s)
Vasos Coronarios/efectos de los fármacos , Estradiol/farmacología , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Receptores Acoplados a Proteínas G/agonistas , Animales , Calcio/metabolismo , Células Cultivadas , Vasos Coronarios/citología , Vasos Coronarios/fisiología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiología , Humanos , Canales de Potasio de Gran Conductancia Activados por el Calcio/metabolismo , Canales de Potasio de Gran Conductancia Activados por el Calcio/fisiología , Relajación Muscular/fisiología , Músculo Liso Vascular/fisiología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/metabolismo , Óxido Nítrico/metabolismo , Receptores de Estrógenos , Receptores Acoplados a Proteínas G/metabolismo , Porcinos , Regulación hacia Arriba/efectos de los fármacos , Vasodilatación/fisiología
10.
Am J Physiol Heart Circ Physiol ; 298(5): H1301-7, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20228257

RESUMEN

The marked sexual dimorphism that exists in human cardiovascular diseases has led to the dogmatic concept that testosterone (Tes) has deleterious effects and exacerbates the development of cardiovascular disease in males. While some animal studies suggest that Tes does exert deleterious effects by enhancing vascular tone through acute or chronic mechanisms, accumulating evidence suggests that Tes and other androgens exert beneficial effects by inducing rapid vasorelaxation of vascular smooth muscle through nongenomic mechanisms. While this effect frequently has been observed in large arteries at micromolar concentrations, more recent studies have reported vasorelaxation of smaller resistance arteries at nanomolar (physiological) concentrations. The key mechanism underlying Tes-induced vasorelaxation appears to be the modulation of vascular smooth muscle ion channel function, particularly the inactivation of L-type voltage-operated Ca(2+) channels and/or the activation of voltage-operated and Ca(2+)-activated K(+) channels. Studies employing Tes analogs and metabolites reveal that androgen-induced vasodilation is a structurally specific nongenomic effect that is fundamentally different than the genomic effects on reproductive targets. For example, 5alpha-dihydrotestosterone exhibits potent genomic-androgenic effects but only moderate vasorelaxing activity, whereas its isomer 5beta-dihydrotestosterone is devoid of androgenic effects but is a highly efficacious vasodilator. These findings suggest that the dihydro-metabolites of Tes or other androgen analogs devoid of androgenic or estrogenic effects could have useful therapeutic roles in hypertension, erectile dysfunction, prostatic ischemia, or other vascular dysfunctions.


Asunto(s)
Andrógenos/fisiología , Tono Muscular/fisiología , Músculo Liso Vascular/fisiología , Testosterona/metabolismo , Testosterona/farmacología , Andrógenos/farmacología , Animales , Femenino , Humanos , Masculino , Tono Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Caracteres Sexuales , Relación Estructura-Actividad , Vasodilatación/efectos de los fármacos
13.
Biol Sex Differ ; 11(1): 48, 2020 08 26.
Artículo en Inglés | MEDLINE | ID: mdl-32843085

RESUMEN

BACKGROUND: Acutely, testosterone (TES) and other androgens are efficacious vasodilators, both in vitro and in vivo; however, their long-term effects on arterial blood pressure (BP) remain unclear. It was hypothesized that endogenous androgens exert long-term anti-hypertensive effects on systemic BP through a combination of genomic and nongenomic effects to enhance vasodilation of the systemic vasculature. METHODS: The long-term effects of endogenous TES and exogenous TES replacement therapy (TRT) on BP were studied in intact (InT) and castrated (CsX) male Sprague-Dawley (SD) and testicular-feminized male (Tfm, androgen receptor defective) rats (12 weeks old). Systolic BP (tail-cuff plethysmography) was determined weekly for 15 weeks in InT-control and CsX rats. Some CsX-SD rats received androgen replacement therapy at 10-15 weeks with TES-enanthate (TRT; 1.75 mg/kg, 2x/week) or DHT-enanthate (DRT; 1.00 mg/kg. 2x/week) and a separate group of CsX-SD rats received losartan-potassium in drinking water (LST, 250 mg/L) for the entire 15 week period. Expression of renin, angiotensinogen (Agt), angiotensin converting enzyme (ACE), and angiotensin II type I receptor (AT1R) mRNA in kidney and aorta were determined by real-time PCR (rt-PCR) and plasma renin levels were determined by radioimmunoassay. RESULTS: There was a progressive rise in BP over 10 weeks in CsX (109 ± 3.3 vs. 143 ± 3.5 mmHg), while BP remained stable in InT-control (109 ± 3.0 vs. 113 ± 0.3). BP gradually declined to normal in CsX-TRT rats (113 ± 1.3), while BP remained elevated in CsX (140 ± 1.2) and normal in InT-control (113 ± 0.3). LST prevented the development of hypertension in CsX at 10 weeks (100 ± 1.5 in CsX + LST vs. 143 ± 3.5 in CsX). During the next 5 weeks with TES-RT, BP declined in CsX-TRT (113 ± 1.3) and remained lower in CsX + LST (99 ± 0.4). DHT-RT reduced BP in CxS to a similar extent. In Tfm, CsX resulted in a similar rise in BP (109 ± 0.7 vs. 139 ± 0.4 mmHg), but TRT reduced BP more rapidly and to a greater extent (106 ± 2.8). rt-PCR of the kidney revealed that CsX increased expression of mRNA for renin (92%), ACE (58%), and AT1R (80%) compared to InT, while TES RT normalized expression of renin, AT1R, and ACE mRNA to levels of InT rats. Plasma renin levels exhibited changes similar to those observed for renin mRNA expression. CONCLUSIONS: This is the first study to examine the long-term effects of endogenous and exogenous androgens on BP in male SD and Tfm rats. These data reveal that endogenous androgens (TES) exert anti-hypertensive effects that appear to involve non-genomic and possibly genomic mechanism(s), resulting in reductions in RAS expression in the kidney and enhanced systemic vasodilation.


Asunto(s)
Andrógenos/metabolismo , Hipertensión/metabolismo , Sistema Renina-Angiotensina/fisiología , Testosterona/análogos & derivados , Andrógenos/farmacología , Animales , Antihipertensivos/uso terapéutico , Aorta/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Hipertensión/tratamiento farmacológico , Riñón/metabolismo , Losartán/uso terapéutico , Masculino , Orquiectomía , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Testosterona/farmacología
14.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1713-23, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19812360

RESUMEN

The risk for cardiovascular disease (CVD) increases with advancing age; however, the age at which CVD risk increases significantly is delayed by more than a decade in women compared with men. This cardioprotection, which women experience until menopause, is presumably due to the presence of ovarian hormones, in particular, estrogen. The purpose of this study was to determine how age and ovarian hormones affect flow-induced vasodilation in the coronary resistance vasculature. Coronary arterioles were isolated from young (6 mo), middle-aged (14 mo), and old (24 mo) intact, ovariectomized (OVX), and ovariectomized + estrogen replaced (OVE) female Fischer-344 rats to assess flow-induced vasodilation. Advancing age impaired flow-induced dilation of coronary arterioles (young: 50 +/- 4 vs. old: 34 +/- 6; % relaxation). Ovariectomy reduced flow-induced dilation in arterioles from young females, and estrogen replacement restored vasodilation to flow. In aged females, flow-induced vasodilation of arterioles was unaltered by OVX; however, estrogen replacement improved flow-induced dilation by approximately 160%. The contribution of nitric oxide (NO) to flow-induced dilation, assessed by nitric oxide synthase (NOS) inhibition with N(G)-nitro-l-arginine methyl ester (l-NAME), declined with age. l-NAME did not alter flow-induced vasodilation in arterioles from OVX rats, regardless of age. In contrast, l-NAME reduced flow-induced vasodilation of arterioles from estrogen-replaced rats at all ages. These findings indicate that the age-induced decline of flow-induced, NO-mediated dilation in coronary arterioles of female rats is related, in part, to a loss of ovarian estrogen, and estrogen supplementation can improve flow-induced dilation, even at an advanced age.


Asunto(s)
Envejecimiento , Circulación Coronaria/efectos de los fármacos , Vasos Coronarios/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Estradiol/administración & dosificación , Terapia de Reemplazo de Estrógeno , Ovariectomía , Vasodilatación/efectos de los fármacos , Factores de Edad , Animales , Arteriolas/efectos de los fármacos , Vasos Coronarios/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Implantes de Medicamentos , Endotelio Vascular/metabolismo , Inhibidores Enzimáticos/farmacología , Femenino , Hidrazinas/farmacología , Indometacina/farmacología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas F344
15.
PLoS One ; 13(1): e0191418, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29360846

RESUMEN

Estrogen exerts protective effects against cardiovascular diseases in premenopausal women, but is associated with an increased risk of both coronary heart disease and stroke in older postmenopausal women. Studies have shown that activation of the G-protein-coupled estrogen receptor 1 (GPER) can cause either relaxation or contraction of arteries. It is highly likely that these dual actions of GPER may contribute to the seemingly paradoxical effects of estrogen in regulating coronary artery function. The objective of this study was to test the hypothesis that activation of GPER enhances agonist-stimulated porcine coronary artery contraction via epidermal growth factor receptor (EGFR) transactivation and its downstream extracellular signal-regulated kinases (ERK1/2) pathway. Isometric tension studies and western blot were performed to determine the effect of GPER activation on coronary artery contraction. Our findings demonstrated that G-1 caused concentration-dependent relaxation of ET-1-induced contraction, while pretreatment of arterial rings with G-1 significantly enhanced ET-1-induced contraction. GPER antagonist, G-36, significantly inhibited both the G-1-induced relaxation effect and G-1-enhanced ET-1 contraction. Gallein, a Gßγ inhibitor, significantly increased G-1-induced relaxation, yet inhibited G-1-enhanced ET-1-mediated contraction. Similarly, inhibition of EGFR with AG1478 or inhibition of Src with phosphatase 2 further increased G-1-induced relaxation responses in coronary arteries, but decreased G-1-enhanced ET-1-induced contraction. Western blot experiments in porcine coronary artery smooth muscle cells (PCASMC) showed that G-1 increased tyrosine phosphorylation of EGFR, which was inhibited by AG-1478. Furthermore, enzyme-linked immunosorbent assays showed that the level of heparin-binding EGF (HB-EGF) released by ET-1 treatment increased two-fold; whereas pre-incubation with G-1 further increased ET-1-induced HB-EGF release to four-fold over control conditions. Lastly, the role of ERK1/2 was determined by applying the MEK inhibitor, PD98059, in isometric tension studies and detecting phospho-ERK1/2 in immunoblotting. PD98059 potentiated G-1-induced relaxation response, but blocked G-1-enhanced ET-1-induced contraction. By western blot, G-1 treatment decreased phospho-ERK1/2, however, in the presence of the adenylyl cyclase inhibitor, SQ22536, G-1 significantly increased ERK1/2 phosphorylation in PCASMC. These data demonstrate that activation of GPER induces relaxation via cAMP as well as contraction via a mechanism involving transactivation of EGFR and the phosphorylation of ERK1/2 in porcine coronary arteries.


Asunto(s)
Vasos Coronarios/fisiología , Receptores ErbB/genética , Receptores de Estrógenos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Células Cultivadas , Vasos Coronarios/efectos de los fármacos , Ciclopentanos/farmacología , Flavonoides/farmacología , Humanos , Técnicas In Vitro , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Modelos Cardiovasculares , Miocitos del Músculo Liso/metabolismo , Quinazolinas/farmacología , Quinolinas/farmacología , Receptores Acoplados a Proteínas G/agonistas , Porcinos , Activación Transcripcional , Tirfostinos/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatación/genética , Vasodilatación/fisiología
16.
J Steroid Biochem Mol Biol ; 178: 65-72, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29113921

RESUMEN

Dehydroepiandrosterone (DHEA), testosterone (TES) and its 5-reduced metabolites induce a nongenomic vasorelaxation in several vascular beds of mammals; similarly these hormones produce systemic hypotensive and antihypertensive responses in normotensive and hypertensive male rats. Thus, it was hypothesized that the antihypertensive response of androgens, whose levels are elevated during gestation, protect against gestational hypertension. An animal model of preeclampsia was induced in female Wistar rats using DOCA-salt-treated pregnant (PT) and normal pregnant (NP) rats. In vivo experiments in conscious rats revealed that bolus intravenous injections of DHEA, TES, 5α- or 5ß-dihydrotestosterone (-DHT) log -1.0 to 2.0µmolk-1min-1, produced substantial transient reductions in arterial blood pressure (BP), without significant changes in heart rate (HR). Mean arterial blood pressure (MAP) was reduced significantly in both groups. PT rats were more sensitive to the antihypertensive responses of androgens than NP. DHEA and 5ß-DHT were the most potent to reduce MAP: 66±07 and 69±2.0mmHg in PT but only 33±0.5 and 35±1.2mmHg in NP rats, respectively. In isolated aortas of PT and NP, the concentration-response curves to each androgen (0.1-100µM) indicated that KCl-induced pre-contraction is more sensitive to all androgens than phenylephrine (Phe) pre-contractions. Notably, 5ß-DHT is the greatest vasorelaxant with KCl-induced contraction than with Phe contraction of both groups, suggesting a preferential blockade on L-VOCCs. TES exhibited minor vasorelaxing effect of aortas pre-contracted with KCl, compared to its precursor DHEA and its 5-reduced metabolites. These data show that these androgens exert acute vasorelaxing effects in vitro and remarkably, reduce the BP in vivo in PT and NP at term pregnancy. Moreover, a deficit in feto-placental androgen production during pregnancy may trigger the development of preeclampsia or gestational hypertension.


Asunto(s)
Andrógenos/farmacología , Antihipertensivos/farmacología , Modelos Teóricos , Preeclampsia/tratamiento farmacológico , Vasodilatación/efectos de los fármacos , Animales , Femenino , Embarazo , Ratas , Ratas Wistar
17.
J Steroid Biochem Mol Biol ; 167: 106-114, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27888135

RESUMEN

Androgens are vasoactive steroids that induce acute vasodilation in a number of isolated vascular beds from different species, but the effects of these hormones on systemic blood pressure (BP) have been studied little. Although it has been reported that androgens exert systemic hypotensive effects through peripheral vasodilation in normotensive rats, there have not been any reports of systemic hypotensive effects of androgens in animals with hypertension. This study was designed to evaluate the acute effects of testosterone (TES) and its 5-reduced metabolites on systemic BP in hypertensive rats and to test the hypothesis that hypotestosteronemia may be involved in the pathogenesis of hypertension. Chronic, indwelling catheters were implanted in carotid artery and jugular vein of 18-21-week-old male spontaneously hypertensive rats (SHR) and normotensive-control Wistar-Kyoto (WKY) rats, for BP recording and drug administration, respectively. Bolus injections of TES, 5α- or 5ß-dihydrotestosterone (5α- and 5ß-DHT), were administrated cumulatively to conscious rats at doses of 0.1-100µmolkg-1min-1. 5ß-DHT was also administrated during the pressor effect of Bay K 8644, an L-type voltage-operated Ca2+ channel (L-VOCC) agonist. In separate experiments, BP of orchidectomized normotensive male WKY and Wistar rats, with or without androgen-replacement therapy, was evaluated weekly for 10 weeks by tail-cuff plethysmography. TES and its metabolites reduced BP in a dose-dependent manner, while heart rate was reduced with some androgens at the highest doses. The hypotensive effects of androgens were markedly greater in SHR, with a rank order potency of: 5ß-DHT>TES>5α-DHT. 5ß-DHT, the most potent antihypertensive androgen, abolished the pressor response to Bay K 8644 in SHR. TES deprivation by orchidectomy increased BP in normotensive WKY and Wistar rats, but this hypertension was prevented by TES replacement therapy. BP responses to androgens are androgen structure-dependent. These data indicate that: 1) androgens play a significant role in the control of BP and may contribute to the pathogenesis of hypertension; 2) blockade of L-VOCC is involved in the antihypertensive effects of androgens, which are non-genomically mediated; and 3) hypotestosteronemia may be a risk factor for hypertension.


Asunto(s)
Andrógenos/farmacología , Antihipertensivos/farmacología , Presión Sanguínea/efectos de los fármacos , Estado de Conciencia/efectos de los fármacos , Hipertensión/patología , Animales , Canales de Calcio Tipo L/metabolismo , Dihidrotestosterona/farmacología , Hipertensión/metabolismo , Masculino , Orquiectomía , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Ratas Wistar , Factores de Riesgo , Testosterona/farmacología
18.
PLoS One ; 12(3): e0173085, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28278256

RESUMEN

Previously, we reported that cAMP/PKA signaling is involved in GPER-mediated coronary relaxation by activating MLCP via inhibition of RhoA pathway. In the current study, we tested the hypothesis that activation of GPER induces coronary artery relaxation via inhibition of RhoA/Rho kinase pathway by cAMP downstream targets, exchange proteins directly activated by cAMP (Epac) as well as PKA. Our results show that Epac inhibitors, brefeldin A (BFA, 50 µM), or ESI-09 (20 µM), or CE3F4 (100 µM), all partially inhibited porcine coronary artery relaxation response to the selective GPER agonist, G-1 (0.3-3 µM); while concurrent administration of BFA and PKI (5 µM), a PKA inhibitor, almost completely blocked the relaxation effect of G-1. The Epac specific agonist, 8-CPT-2Me-cAMP (007, 1-100 µM), induced a concentration-dependent relaxation response. Furthermore, the activity of Ras-related protein 1 (Rap1) was up regulated by G-1 (1 µM) treatment of porcine coronary artery smooth muscle cells (CASMCs). Phosphorylation of vasodilator-stimulated phosphoprotein (p-VASP) was elevated by G-1 (1 µM) treatment, but not by 007 (50 µM); and the effect of G-1 on p-VASP was blocked by PKI, but not by ESI-09, an Epac antagonist. RhoA activity was similarly down regulated by G-1 and 007, whereas ESI-09 restored most of the reduced RhoA activity by G-1 treatment. Furthermore, G-1 decreased PGF2α-induced p-MYPT1, which was partially reversed with either ESI-09 or PKI; whereas, concurrent administration of ESI-09 and PKI totally prevented the inhibitory effect of G-1. The inhibitory effects of G-1 on p- MLC levels in CASMCs were mostly restored by either ESI-09 or PKI. These results demonstrate that activation of GPER induces coronary artery relaxation via concurrent inhibition of RhoA/Rho kinase by Epac/Rap1 and PKA. GPER could be a potential drug target for preventing and treating cardiovascular diseases.


Asunto(s)
Vasos Coronarios/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Receptores de Estrógenos/metabolismo , Proteínas de Unión al GTP rap1/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Células Cultivadas , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Ciclopentanos/farmacología , Factores de Intercambio de Guanina Nucleótido/antagonistas & inhibidores , Factores de Intercambio de Guanina Nucleótido/metabolismo , Hidrazonas/farmacología , Isoxazoles/farmacología , Músculo Liso Vascular/citología , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/metabolismo , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Fosforilación/efectos de los fármacos , Quinolinas/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Transducción de Señal/efectos de los fármacos , Porcinos , Tionucleótidos/farmacología , Proteínas de Unión al GTP rap1/antagonistas & inhibidores , Proteínas de Unión al GTP rap1/genética
19.
Cardiovasc Res ; 63(2): 338-46, 2004 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-15249192

RESUMEN

OBJECTIVES: Endothelial nitric oxide synthase (eNOS) activation/deactivation is associated with cyclic depalmitoylation/repalmitoylation of specific Cys residues. The mechanism of depalmitoylation has been identified recently, but repalmitoylation remains undefined. We hypothesized that long chain fatty acyl CoA synthetase (LCFACoAS) modulates endothelial nitric oxide synthase repalmitoylation by limiting palmitoyl CoA availability. METHODS: Human coronary endothelial cells were treated with triacsin-C, an inhibitor of long chain fatty acyl CoA synthetase, for 24 h. Media nitrite accumulation, eNOS activity, and eNOS palmitoylation were measured. Methacholine-induced NO synthesis or vascular relaxation were measured in endothelium-intact rat aortae in the presence and absence of triacsin-C. RESULTS: Triacsin-C significantly reduced incorporation of [3H] palmitate into immunoreactive endothelial nitric oxide synthase and over a concentration range of 0.1 to 10 microM, increased media nitrite accumulations 2- to 2.5-fold over baseline. Total in vitro catalytic activity of nitric oxide synthase in triacsin-C treated cells did not differ significantly from control. Triacsin-C significantly increased methacholine-induced NO synthesis in the isolated rat aorta, and significantly enhanced methacholine-induced relaxation of rat aortic rings. CONCLUSIONS: These data are consistent with the interpretation that inhibition of palmitoylation increases endothelial nitric oxide synthase activity without changing endothelial nitric oxide synthase expression, suggesting that inhibiting palmitoylation increases the catalytically active fraction of endothelial nitric oxide synthase.


Asunto(s)
Colina/análogos & derivados , Coenzima A Ligasas/antagonistas & inhibidores , Vasos Coronarios , Endotelio Vascular/metabolismo , Hipertensión/metabolismo , Óxido Nítrico/metabolismo , Triazenos/farmacología , Animales , Células Cultivadas , Colina/farmacología , Citoplasma/enzimología , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo III , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley
20.
Oecologia ; 38(2): 203-216, 1979 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28308890

RESUMEN

Dusky-footed woodrats (Neotoma fuscipes) and desert woodrats (N. lepida) experience pronounced seasonal variations in the aridity of their habitats. The effects of seasonal aridity upon the water conserving abilities of these species were assessed through measurements of water conserving abilities and kidney structure of animals captured in summer and winter, and through measurements of animals' abilities to acclimate to differing water availabilities in the laboratory.Urine concentrating ability was the water conserving mechanism most responsive to changes in the availability of water. Summer and summeracclimated N. fuscipes (431.7 and 459.4 mEqCl-/1) demonstrated urine Cl- concentrating abilities substantially greater than those of winter and winter-acclimated N. fuscipes (245.7 and 337.4 mEqCl-/1). Summer, winter-acclimated, and winter N. lepida exhibited urine Cl- concentrations equivalent to those of winter N. fuscipes; summer-acclimated N. lepida exhibited markedly greater values (466.7 mEqCl-/l) equivalent to those of summer and summer-acclimated N. fuscipes.Measurements of relative thicknesses of renal cortex and medulla yielded no significant differences among the experimental groups of N. fuscipes and N. lepida, thus suggesting that both species possess equal abilities to concentrate urine. These data are confirmed by urine concentrations of summer-acclimated animals of both species.Water conserving abilities of both species correlate well with climatic and dietary plant water content data. Thus, during the dry, warm summer months (when plant moisture is reduced) N. fuscipes conserves water mainly through increased urine concentration. The laboratory acclimation data and differences between summer and winter animals strongly suggest that N. fuscipes undergoes an acclimatization to the seasonal aridity which increases gradually during spring and peaks in late summer, thus enabling this water-dependent species to exist on reduced water requirements. The uniformly low water conserving abilities of winter-acclimated, winter, and summer N. lepida physiologically verify the previous reports that this species satisfies its water requirements through utilization of succulent cactus, thereby avoiding the stress of summer aridity in its habitat.The fact that both species exhibit equal capacities to conserve water indicates that the much greater geographic distribution of N. lepida is not the result of differences in physiological water conserving abilities, but instead may be the result of specific physiological adaptation by N. lepida to utilization of cactus and other plants containing noxious or toxic compounds.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA