Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

País de afiliación
Intervalo de año de publicación
1.
Exp Physiol ; 108(3): 412-419, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36651722

RESUMEN

NEW FINDINGS: What is the central question of this study? Do alterations in thyroid status affect haemodynamic parameters and echocardiographic measurements in the rat postnatal heart, and calcium handling, contractility, relaxation and cardiolipin content in isolated rat cardiomyocytes? What is the main finding and its importance? An imbalance in phospholipids of the mitochondrial membrane such as cardiolipin is related to defects in mitochondrial function. T3 -dependent cardiolipin signals contribute to the maintenance of mitochondrial homeostasis and involve Ca2+ handling, this pathway being more important in hypothyroidism. ABSTRACT: The objective of this study was to evaluate whether alterations in thyroid status affect (1) haemodynamic parameters and echocardiographic measurements in the rat postnatal heart, and (2) calcium handling, contractility, relaxation and cardiolipin content in isolated rat cardiomyocytes. Sprague-Dawley rats aged 2 months treated with T3 (hyperthyroid, 20 µg/100 g body weight) or 0.02% methimazole (hypothyroid, w/v) for 28 days. Heart function was evaluated by echocardiography. Measurements of mean arterial pressure (MAP), heart rate, Ca2+ transients, cardiomyocyte shortening, number of spontaneous contractions per minute and cardiolipin (CL) content were performed. Thyroid disorders were associated with changes in pacemaker activity without modifications of MAP. Thyroid disorder induced changes in left ventricular diameter which were correlated with modifications of cardiac contractility (altered cell shortening and sarcoplasmic reticulum Ca2+ content). Endocrine disorders altered cardiomyocyte relaxation (reduction in the time to 50% re-lengthening and the time to 50% Ca2+ decay). Thyroid disorder increased the number of spontaneous contractions per minute (an index of pro-arrhythmogenic behaviour). CL content was increased only in hypothyroid rats. Changes in CL content, CL composition and CL-protein interaction in mitochondria from hypothyroid animals are responsible for alterations of contractile and relaxation cardiac function. This mechanism may be not be involved in T3 -treated rats. Maintenance of euthyroidism is of crucial importance to preserve cardiac performance. An imbalance in relation to phospholipids of the mitochondrial membrane such as CL is related to defects in mitochondrial function. T3 -dependent CL signals contribute to the maintenance of mitochondrial homeostasis and involve Ca2+ handling, this pathway being more important in hypothyroidism.


Asunto(s)
Cardiolipinas , Hipotiroidismo , Ratas , Animales , Cardiolipinas/metabolismo , Calcio/metabolismo , Ratas Sprague-Dawley , Hormonas Tiroideas/metabolismo , Miocitos Cardíacos/metabolismo , Contracción Miocárdica , Retículo Sarcoplasmático/metabolismo
2.
J Cardiovasc Pharmacol ; 78(6): 784-791, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524257

RESUMEN

ABSTRACT: The aim of the present work was to examine whether metabolic syndrome-like conditions in rats with fructose (F) overload modify the cardiotoxic effects induced by doxorubicin (DOX) and whether the treatment altered the expression of P-gp, breast cancer resistance protein, and organic cation/carnitine transporters in the heart. Male Sprague-Dawley rats received either tap water (control group [C]; n = 16) or water with F 10% wt/vol (n = 16) during 8 weeks. Three days before being killed, the animals received a single dose of DOX (6 mg/kg, ip, md) (C-DOX and F-DOX groups) or vehicle (VEH; ISS 1 mL/kg BW; ip) (C-VEH and F-VEH groups) (n = 8 per group). F overload enhanced thiobarbituric acid-reactive substance levels in the left ventricle, and DOX injection further increased those values. DOX did not alter thiobarbituric acid-reactive substance production in C animals. DOX caused a decrease of 30% in the ejection fraction and a nearly 40% reduction in the fractional shortening in F animals, but not in C rats. Cardiac tissue levels of P-gp decreased by about 30% in F rats compared with the C groups. DOX did not modify cardiac P-gp expression. Breast cancer resistance protein and organic cation/carnitine transporter (OCTN 1/2/3) protein levels did not change with either F or DOX. It is suggested that DOX could cause greater cardiotoxicity in rats receiving F, probably due to enhanced cardiac lipid peroxidation and lower expression of cardiac P-gp. These results support the hypothesis that the cardiotoxicity of DOX could be increased under metabolic syndrome-like conditions or in other health disorders that involve cardiovascular risk factors.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Antibióticos Antineoplásicos , Doxorrubicina , Cardiopatías/inducido químicamente , Síndrome Metabólico/complicaciones , Miocardio/metabolismo , Estrés Oxidativo , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Animales , Cardiotoxicidad , Modelos Animales de Enfermedad , Cardiopatías/metabolismo , Cardiopatías/patología , Cardiopatías/fisiopatología , Peroxidación de Lípido , Masculino , Síndrome Metabólico/metabolismo , Miocardio/patología , Proteínas de Transporte de Catión Orgánico/genética , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas Sprague-Dawley , Función Ventricular Izquierda/efectos de los fármacos
3.
J Cardiovasc Pharmacol ; 73(5): 290-300, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31082960

RESUMEN

Erythropoietin (EPO) has been linked to cardioprotective effects. However, its effects during the aging process are little known. We investigated the effect of EPO administration on hemodynamic parameters, cardiac function, oxidative damage, and erythropoietin receptor (EPOR) expression pattern in the hypovolemic state. EPO was administered (1000 IU/kg/3 days) and then acute hemorrhage (20% blood loss) was induced in young and adult rats. There was no difference in plasmatic EPO in either age group. The hemodynamic basal condition was similar, without alterations in renal function and hematocrit, in both age groups. After bleeding, both EPO-treated age groups had increased blood pressure at the end of the experimental protocol, being greater in adult animals. EPO attenuated the tachycardic effect. Ejection fraction and fractional shortening were higher in adult EPO-treated rats subjected to hemorrhage. In the left ventricle, young and adult EPO-treated rats subjected to bleeding showed an increased EPOR expression. A different EPOR expression pattern was observed in the adult right atrial tissue, compared with young animals. EPO treatment decreased oxidative damage to lipids in both age groups. EPO treatment before acute hemorrhage improves cardiovascular function during the aging process, which is mediated by different EPOR pattern expression in the heart tissue.


Asunto(s)
Sistema Cardiovascular/efectos de los fármacos , Epoetina alfa/administración & dosificación , Hematínicos/administración & dosificación , Hemodinámica/efectos de los fármacos , Hemorragia/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Factores de Edad , Animales , Sistema Cardiovascular/metabolismo , Sistema Cardiovascular/fisiopatología , Modelos Animales de Enfermedad , Hemorragia/metabolismo , Hemorragia/fisiopatología , Peroxidación de Lípido/efectos de los fármacos , Masculino , Miocardio/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Receptores de Eritropoyetina/agonistas , Receptores de Eritropoyetina/metabolismo
4.
Eur J Nutr ; 55(1): 33-43, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25519003

RESUMEN

PURPOSE: During the postnatal stage, cardiovascular nitric oxide (NO) system and caveolins (cav) may be regulated differentially in response to hypovolemic state induced by water restriction. Our aim was to examine the effects of water restriction on NO synthases (NOS) and cav in the atria, ventricle and aorta of growing rats. METHODS: Male Sprague-Dawley rats aged 25 and 50 days were divided into (n = 15): WR: water restriction 3 days; WAL: water ad libitum 3 days. Systolic blood pressure, NOS activity and NOS/cav protein levels were measured. RESULTS: Dehydration induced a larger increase in SBP in WR25 group. Ventricular NOS activity, endothelial NOS (eNOS) and neuronal isoform (nNOS) of WR25 pups were increased, and both cav were decreased. In the WR50 group, NOS activity remained unchanged. In the atria, NOS activity, eNOS and nNOS decreased in WR25 associated with increased cav-1; in the WR50 group, NOS activity was increased without changes in NOS isoforms. In the aorta of WR25, NOS activity and inducible NOS (iNOS) were decreased; NOS activity was unchanged in WR50, despite the decreased levels of eNOS and increased iNOS, cav-1 and cav-3. CONCLUSIONS: NO system adjustments in cardiovascular system under osmotic stress in vivo depend on postnatal age, being eNOS and nNOS, the isoforms that determine NOS activity in cardiac tissue in 25-day-old pups. Changes in cav abundance during hypovolemic state may contribute to age-related NO production.


Asunto(s)
Sistema Cardiovascular/metabolismo , Caveolina 1/metabolismo , Caveolina 3/metabolismo , Deshidratación , Óxido Nítrico Sintasa de Tipo I/metabolismo , Animales , Presión Sanguínea , Caveolina 1/genética , Caveolina 3/genética , Endotelio/metabolismo , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Hemodinámica , Hipovolemia/metabolismo , Masculino , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Presión Osmótica , Ratas , Ratas Sprague-Dawley , Sustancias Reactivas al Ácido Tiobarbitúrico
5.
Eur J Nutr ; 53(1): 287-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23625137

RESUMEN

PURPOSE: Aquaporin-1 (AQP1) is the predominant water channel in the heart, linked to cardiovascular homeostasis. Our aim was to study cardiovascular AQP1 distribution and protein levels during osmotic stress and subsequent hydration during postnatal growth. METHODS: Rats aged 25 and 50 days were divided in: 3d-WR: water restriction 3 days; 3d-WAL: water ad libitum 3 days; 6d-WR+ORS: water restriction 3 days + oral rehydration solution (ORS) 3 days; and 6d-WAL: water ad libitum 6 days. AQP1 was evaluated by immunohistochemistry and western blot in left ventricle, right atrium and thoracic aorta. RESULTS: Water restriction induced a hypohydration state in both age groups (40 and 25 % loss of body weight in 25- and 50-day-old rats, respectively), reversible with ORS therapy. Cardiac AQP1 was localized in the endocardium and endothelium in both age groups, being evident in cardiomyocytes membrane only in 50-day-old 3d-WR group, which presented increased protein levels of AQP1; no changes were observed in the ventricle of pups. In vascular tissue, AQP1 was present in the smooth muscle of pups; in the oldest group, it was found in the endothelium, increasing after rehydration in smooth muscle. No differences were observed between control groups 3d-WAL and 6d-WAL of both ages. CONCLUSION: Our findings suggest that cardiovascular AQP1 can be differentially regulated in response to hydration status in vivo, being this response dependent on postnatal growth. The lack of adaptive mechanisms of mature animals in young pups may indicate an important role of this water channel in maintaining fluid balance during hypovolemic state.


Asunto(s)
Acuaporina 1/metabolismo , Miocitos Cardíacos/metabolismo , Privación de Agua/fisiología , Agua/administración & dosificación , Glándulas Suprarrenales/metabolismo , Animales , Aorta Torácica/metabolismo , Composición Corporal , Peso Corporal , Endotelio/metabolismo , Atrios Cardíacos/metabolismo , Ventrículos Cardíacos/metabolismo , Homeostasis , Hipovolemia/metabolismo , Hipovolemia/patología , Hipovolemia/terapia , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Equilibrio Hidroelectrolítico/fisiología
6.
Clin Sci (Lond) ; 125(11): 521-32, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23642207

RESUMEN

We have reported previously that centrally applied ET (endothelin)-1 and ET-3 induce either choleresis or cholestasis depending on the dose. In the present study, we sought to establish the role of these endothelins in the short-term peripheral regulation of bile secretion in the rat. Intravenously infused endothelins induced significant choleresis in a dose-dependent fashion, ET-1 being more potent than ET-3. Endothelins (with the exception of a higher dose of ET-1) did not affect BP (blood pressure), portal venous pressure or portal blood flow. ET-1 and ET-3 augmented the biliary excretion of bile salts, glutathione and electrolytes, suggesting enhanced bile acid-dependent and -independent bile flows. ET-induced choleresis was mediated by ET(B) receptors coupled to NO and inhibited by truncal vagotomy, atropine administration and capsaicin perivagal application, supporting the participation of vagovagal reflexes. RT (reverse transcription)-PCR and Western blot analysis revealed ETA and ET(B) receptor expression in the vagus nerve. Endothelins, through ET(B) receptors, augmented the hepatocyte plasma membrane expression of Ntcp (Na⁺/taurocholate co-transporting polypeptide; Slc10a1), Bsep (bile-salt export pump; Abcb11), Mrp2 (multidrug resistance protein-2; Abcc2) and Aqp8 (aquaporin 8). Endothelins also increased the mRNAs of these transporters. ET-1 and ET-3 induced choleresis mediated by ET(B) receptors coupled to NO release and vagovagal reflexes without involving haemodynamic changes. Endothelin-induced choleresis seems to be caused by increased plasma membrane translocation and transcriptional expression of key bile transporters. These findings indicate that endothelins are able to elicit haemodynamic-independent biological effects in the liver and suggest that these peptides may play a beneficial role in pathophysiological situations where bile secretion is impaired.


Asunto(s)
Colestasis/inducido químicamente , Endotelina-1/farmacología , Endotelina-3/farmacología , Óxido Nítrico/fisiología , Receptor de Endotelina B/fisiología , Nervio Vago/efectos de los fármacos , Animales , Bilis/metabolismo , Presión Sanguínea/efectos de los fármacos , Colagogos y Coleréticos/farmacología , Colestasis/metabolismo , Hemodinámica/efectos de los fármacos , Óxido Nítrico/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor de Endotelina B/metabolismo , Reflejo/efectos de los fármacos , Flujo Sanguíneo Regional/efectos de los fármacos , Vagotomía , Nervio Vago/metabolismo , Nervio Vago/fisiología
7.
Mol Cell Biochem ; 359(1-2): 169-76, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21842376

RESUMEN

Hemorrhage (H) is associated with a left ventricular (LV) dysfunction. However, the diastolic function has not been studied in detail. The main goal was to assess the diastolic function both during and 120 min after bleeding, in the absence and in the presence of L-NAME. Also, the changes in mRNA and protein expression of nitric oxide synthase (NOS) isoforms were determined. New Zealand rabbits were divided into three groups: Sham group, H group (hemorrhage 20% blood volume), and H L-NAME group (hemorrhage treated with L-NAME). We evaluated systolic and diastolic ventricular functions in vivo and in vitro (Langendorff technique). Hemodynamic parameters and LV function were measured before, during, and at 120 min after bleeding. We analyzed the isovolumic relaxation using t ½ in vivo (closed chest). After that, hearts were excised and perfused in vitro to measure myocardial stiffness. Samples were frozen to measure NOS mRNA and protein expression. The t½ increased during bleeding and returned to basal values 120 min after bleeding. L-NAME blunted this effect. Data from the H group revealed a shift to the left in the LV end diastolic pressure-volume curve at 120 min after bleeding, which was blocked by L-NAME. iNOS and nNOS protein expression and mRNA levels increased at 120 min after the hemorrhage. Acute hemorrhage induces early and transient isovolumic relaxation impairment and an increase in myocardial stiffness 120 min after bleeding. L-NAME blunted the LV dysfunction, suggesting that NO modulates ventricular function through iNOS and nNOS isoforms.


Asunto(s)
Diástole , Choque Hemorrágico/fisiopatología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Animales , Diástole/efectos de los fármacos , Diástole/fisiología , Corazón/fisiopatología , Hemorragia , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo I , Óxido Nítrico Sintasa de Tipo II , Óxidos de Nitrógeno , Conejos , Choque Hemorrágico/complicaciones , Disfunción Ventricular Izquierda/enzimología , Disfunción Ventricular Izquierda/etiología
8.
Eur J Nutr ; 50(8): 657-64, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21404082

RESUMEN

AIM OF STUDY: We have assessed the influence of water restriction stress on the nitric oxide (NO) synthase in heart and aorta tissues in young 2-month-old and middle-aged 12-month-old rats. METHODS: Animals were divided into control and 24- and 72-h water-deprived groups. We evaluated systolic blood pressure (SBP), biochemical parameters, nitrate and nitrite urinary excretion (UNOx), NADPH-diaphorase activity, and protein levels of NOS in the right atria, left ventricle, and thoracic aorta tissues. RESULTS: Water restriction during 72 h increased SBP (16%) in 2-month-old rats but decreased it after 24 and 72 h (9 and 15%, respectively) in 12-month-old rats. Atria, aorta endothelium, and smooth muscle NOS activity increased (32, 63, and 88%, respectively) only after 72 h of water restriction in 2-month-old rats. It also increased not only after 72 h but also after 24 h in atria (27 and 18%, respectively) and in ventricle (39 and 67%, respectively) in 12-month-old rats. Meanwhile, in this group's aorta smooth muscle, the enzyme activity decreased (16 and 7%, respectively). A major difference seen between ages was the changes in UNOx excretion, which decreased in the younger in 24 and 72 h (47 and 81%, respectively) and increased in the middle-aged rats (193 and 389%, respectively). Water restriction did not change cardiovascular endothelial and neuronal NOS protein levels in any group. CONCLUSION: NO pathways could contribute to the development of age-related cardiovascular adaptation to volume depletion induced by water restriction.


Asunto(s)
Envejecimiento/patología , Ventrículos Cardíacos/enzimología , Hipovolemia/patología , Óxido Nítrico Sintasa/metabolismo , Agua/fisiología , Adaptación Fisiológica , Animales , Aorta Torácica/enzimología , Presión Sanguínea , Endotelio Vascular/enzimología , Hipovolemia/enzimología , Masculino , Modelos Animales , NADPH Deshidrogenasa/metabolismo , Nitratos/orina , Nitritos/orina , Ratas , Ratas Sprague-Dawley , Estrés Fisiológico
9.
J Mol Endocrinol ; 66(3): 207-221, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33640872

RESUMEN

Recent studies have provided evidence that triiodothyronine (T3) might play an effective role in the recovery of ischemic myocardium, through the preservation of mitochondrial function and the improvement of energy substrate metabolism. To this respect, it has been suggested that T3 could activate AMP-activated protein kinase (AMPK), the cellular 'fuel-gauge' enzyme, although its role has yet to be elucidated. The aim of the present study was to investigate the effects produced by acute treatment with T3 (60 nM) and the pharmacological inhibition of AMPK by compound C on isolated rat left atria subjected to 75 min simulated ischemia-75 min reperfusion. Results showed that T3 increased AMPK activation during simulated ischemia-reperfusion, while compound C prevented it. At the end of simulated reperfusion, acute T3 treatment increased contractile function recovery and cellular viability conservation. Mitochondrial ultrastructure was better preserved in the presence of T3 as well as mitochondrial ATP production rate and tissue ATP content. Calcium retention capacity, a parameter widely used as an indicator of the resistance of mitochondrial permeability transition pore (MPTP) to opening, and GSK-3ß phosphorylation, a master switch enzyme that limits MPTP opening, were increased by T3 administration. All these beneficial effects exerted by T3 acute treatment were prevented when compound C was co-administrated. The present study provided original evidence that T3 enhances intrinsic activation of AMPK during myocardial ischemia-reperfusion, being this enzyme involved, at least in part, in the protective effects exerted by T3, contributing to mitochondrial structure and function preservation, post-ischemic contractile recovery and conservation of cellular viability.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Cardiotónicos/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/enzimología , Miocardio/enzimología , Miocardio/patología , Triyodotironina/uso terapéutico , Adenosina Trifosfato/metabolismo , Animales , Calcio/metabolismo , Cardiotónicos/farmacología , Supervivencia Celular/efectos de los fármacos , Diástole/efectos de los fármacos , Femenino , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Atrios Cardíacos/ultraestructura , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/ultraestructura , Contracción Miocárdica/efectos de los fármacos , Daño por Reperfusión Miocárdica/fisiopatología , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Sístole/efectos de los fármacos , Triyodotironina/farmacología
10.
Am J Physiol Heart Circ Physiol ; 298(3): H778-86, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19783776

RESUMEN

The objective was to study atrial natriuretic peptide (ANP) effects on mean arterial pressure (MAP) and cardiovascular nitric oxide (NO) system in spontaneously hypertensive rats (SHRs), investigating the receptors and signaling pathways involved. In vivo, SHRs and Wistar-Kyoto (WKY) rats were infused with saline (0.05 ml/min) or ANP (0.2 microg.kg(-1).min(-1)) for 1 h. MAP and nitrites and nitrates excretion (NOx) were determined. NO synthase (NOS) activity and endothelial (eNOS), neuronal (nNOS) and inducible (iNOS) NOS expression were measured in the heart and aorta. In vitro, heart and aortic NOS activity induced by ANP was determined in the presence of iNOS and nNOS inhibitors, natriuretic peptide receptor (NPR)-A/B blocker, G(i) protein, and calmodulin inhibitors. As a result, ANP diminished MAP and increased NOx in both groups. Cardiovascular NOS activity was higher in SHRs than in WKY rats. ANP increased NOS activity, but the activation was lower in SHRs than in WKY rats. ANP had no effect on NOS isoform expression. NOS activity induced by ANP was not modified by iNOS and nNOS inhibitors. NPR-A/B blockade blunted NOS stimulation via ANP in ventricle and aorta but not in atria. Cardiovascular NOS response to ANP was reduced by G(i) protein and calmodulin inhibitors in both groups. In conclusion, in atria, ventricle, and aorta, ANP interacts with NPR-C receptors, activating Ca(2+)-calmodulin eNOS through G(i) protein. In ventricle and aorta, NOS activation also involves NPR-A/B. The NOS response to ANP was impaired in heart and aorta of SHRs. The impaired NO-system response to ANP in hypertensive animals, involving alterations in the signaling pathway, could participate in the maintenance of high blood pressure in this model of hypertension.


Asunto(s)
Factor Natriurético Atrial/fisiología , Sistema Cardiovascular/fisiopatología , Hipertensión/fisiopatología , Óxido Nítrico/fisiología , Animales , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Masculino , Miocardio/metabolismo , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas WKY , Transducción de Señal/fisiología
11.
Br J Nutr ; 104(3): 382-9, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20338072

RESUMEN

We had previously shown that prenatal exposure to Zn-deficient diets induces an increase in blood pressure and impairs renal function in adult rats. The aim of the present study was to investigate if moderate Zn restriction during early growth periods, fetal life and lactation would induce impairment in the vascular and renal NO system and alterations in plasma lipid profile. We also investigated if these effects persisted into adult life, even when a Zn-replete diet was provided after weaning. Pregnant rats were fed control (30 parts per million (ppm)) or low (8 ppm) Zn diets throughout gestation up to weaning. Afterwards, male offspring from low-Zn mothers were assigned to low- or control-Zn diets during 60 d. Male offspring from control mothers were fed a control diet. Animals exposed to Zn restriction showed low birth weight, increased systolic blood pressure and serum TAG levels, and decreased glomerular filtration rate in adulthood. Zn restriction induced a decrease in vascular and renal NO synthase activity and a reduced expression of the endothelial NO synthase isoform in aorta. A control-Zn diet during post-weaning growth returned TAG levels to normal but was unsuccessful in normalising systolic blood pressure, glomerular filtration rate or NO system activity in Zn-deficient offspring. Zn restriction during fetal life, lactation and/or post-weaning growth induced alterations in the vascular and renal NO system and in lipid metabolism that could contribute to the programming of hypertension and renal dysfunction in adulthood.


Asunto(s)
Presión Sanguínea/efectos de los fármacos , Endotelio Vascular/metabolismo , Óxido Nítrico Sintasa/metabolismo , Efectos Tardíos de la Exposición Prenatal , Fenómenos Fisiologicos de la Nutrición Prenatal , Zinc/deficiencia , Animales , Animales Recién Nacidos , Peso al Nacer , Dieta , Femenino , Tasa de Filtración Glomerular/efectos de los fármacos , Crecimiento , Lactancia , Masculino , Embarazo , Efectos Tardíos de la Exposición Prenatal/sangre , Efectos Tardíos de la Exposición Prenatal/enzimología , Isoformas de Proteínas , Ratas , Ratas Wistar , Triglicéridos/sangre , Destete , Zinc/farmacología
12.
Am J Hypertens ; 21(4): 377-81, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18292759

RESUMEN

Alterations in autonomic control and myocardial nitric-oxide (NO) production are likely linked to the development and progression of heart dysfunction. By focusing on heart rate, the complexity of the actions of NO at distinct levels throughout the autonomic nervous system and its relationship with other regulators can be demonstrated. Given the multiple and opposing actions of NO on cardiac control, it is difficult to interpret a response after a global intervention in the NO system. The diversity of intracellular pathways activated by NO, and their differing sensitivities to different levels of NO, might account for some aspects of reported specific but opposite effects. We discuss factors that might contribute to this diversity of actions. A proper elucidation of the effects of NO on metabolic pathways and on energy generation could lead to novel therapeutic strategies aimed at the early treatment of heart dysfunction.


Asunto(s)
Frecuencia Cardíaca/fisiología , Mitocondrias Cardíacas/metabolismo , Contracción Miocárdica/fisiología , Óxido Nítrico/biosíntesis , Animales , Sistema Nervioso Autónomo/fisiología , Humanos
13.
Regul Pept ; 151(1-3): 130-4, 2008 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-18586055

RESUMEN

UNLABELLED: Atrial natriuretic peptide (ANP) induces activation of nitric oxide-synthase (NOS). AIMS: to identify the isoform of NOS involved in ANP effects, to study whether ANP modifies NOS expression and to investigate the signaling pathways and receptors involved in NOS stimulation. NOS activation induced by ANP would be mediated by endothelial NOS (eNOS) since neuronal or inducible NOS inhibition did not alter ANP effect. The peptide induced no changes in eNOS protein expression. NOS activity stimulated by ANP, in the kidney, aorta and left ventricle, was partially abolished by the NPR-A/B antagonist, as well as PKG inhibition, but no difference in atria was observed. 8-Br-cGMP partially mimicked the effect of ANP on NOS in all tissues. NOS stimulation by ANP in atria disappeared when G protein was inhibited, but this effect was partial in the other tissues. Calmodulin antagonist abolished NOS stimulation via ANP. Inhibition of the PLC, PKC or PI3 kinase/Akt pathway failed to alter NOS activation induced by ANP. ANP would activate eNOS in the aorta, heart and kidney without modifying the expression of the enzyme. ANP would interact with NPR-C coupled via G proteins leading to the activation of Ca(2+)-calmodulin-dependent NOS in atria; while in ventricle, aorta and kidney, ANP could also interact with NPR-A/B, increasing cGMP, which in turns activates PKG to stimulate eNOS.


Asunto(s)
Factor Natriurético Atrial/farmacología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Animales , Aorta/efectos de los fármacos , Aorta/enzimología , Factor Natriurético Atrial/fisiología , Endotelio/efectos de los fármacos , Endotelio/enzimología , Activación Enzimática/efectos de los fármacos , Guanidinas/farmacología , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/enzimología , Riñón/efectos de los fármacos , Riñón/enzimología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Ratas , Ratas Wistar , Transducción de Señal/efectos de los fármacos
14.
Vascul Pharmacol ; 49(1): 19-25, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18502184

RESUMEN

AIM: To examine the effect of nitric oxide (NO) on the expression and/or localization of inner medulla collecting duct aquaporin-2 water channel (AQP2) in young and adult hemorrhaged anesthetized rats. METHODS: Rats of 2 (young) and 12 mo (adult) old (n=15) were divided into: Sham animals with and without NG-nitro-l-arginine methyl ester (L-NAME) treatment (S L-NAME and S); hemorrhaged animals (20% blood loss) with and without L-NAME (H L-NAME and H). Mean arterial pressure (MAP) was continuously monitored and AQP2 expression and inmunolocalization were evaluated at 120 min after bleeding. RESULTS: L-NAME blunted the hypotension induced by hemorrhage at 120 min in young (106+/-2 mm Hg) and adult (103+/-4 mm Hg) rats. AQP2 expression increased after bleeding in young (from 22 to 50 densitometric units) and adult rats (from 15 to 30 densitometric units). Pretreatment with L-NAME enhanced this effect, being this rise lower in adult than young animals (young: 318%, adult: 233%). Electron microscopy showed that AQP2 labeling increased after withdrawal, being the number of gold particles smaller in adult than young animals in the inner medulla. L-NAME enhanced this effect. CONCLUSION: NOS activity decreases AQP2 expression/traffick in the inner collecting duct principal cells in response to hemorrhage and this effect is lower with aging.


Asunto(s)
Envejecimiento/fisiología , Acuaporina 2/metabolismo , Hipovolemia/metabolismo , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Óxido Nítrico/fisiología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Western Blotting , Inhibidores Enzimáticos/farmacología , Hemorragia/metabolismo , Hemorragia/fisiopatología , Inmunohistoquímica , Masculino , Microscopía Inmunoelectrónica , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Agua/metabolismo
15.
J Nephrol ; 21(1): 93-8, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18264941

RESUMEN

BACKGROUND: Salt delivery to the distal nephron and sodium reabsorption in this segment are considered critical factors that modulate kaliuresis in chronic kidney disease (CKD). Amiloride, a drug that blocks Na(+) reabsorption in the distal nephron, can help to assess the role of Na+ transport in this segment on the kaliuresis of CKD patients. METHODS: A bolus of amiloride (1 mg/kg body weight) followed by an intravenous infusion (1 mg/kg body weight per hour) was administered to 6 normal subjects and 10 patients with CKD undergoing water diuresis. Serum and urine electrolytes were measured. Glomerular filtration rate (GFR) was measured with clearance of (125)I-iodothalamate. RESULTS: Normal subjects and CKD patients had a control fractional excretion of potassium (FE(K)(+)) of 26% +/- 11% and 126% +/- 28%, respectively; the corresponding FE(Na)(+) was 2.3% +/- 0.8% and 15% +/- 3%. In response to amiloride, FE(Na)(+)increased significantly to 3.5% +/- 0.6% and 20% +/- 3% in normal and CKD subjects, respectively, and FE(K)(+) decreased significantly to 6.5% +/- 0.6% and 39% +/- 8%, respectively. Amiloride-sensitive and amiloride-insensitive kaliuresis in normal subjects were 71.4% and 28.6%, respectively; the corresponding values for CKD patients were 73% and 27%, respectively. Urine output correlated positively with kaliuresis in CKD. CONCLUSIONS: The very high FEK+ observed in CKD occurs in the absence of hyperkalemia and is largely amiloride-sensitive; therefore maintenance of potassium balance by the kidney in CKD is mostly dependent on sodium reabsorption through channels along the distal nephron. The high urinary flow of CKD further promotes potassium excretion.


Asunto(s)
Amilorida/farmacología , Enfermedades Renales/metabolismo , Potasio/orina , Adulto , Anciano , Enfermedad Crónica , Canales Epiteliales de Sodio/fisiología , Femenino , Humanos , Masculino , Persona de Mediana Edad
16.
Vascul Pharmacol ; 44(6): 417-26, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16697268

RESUMEN

AIM: Nitric oxide has been implicated in the cardiovascular adaptation to hemorrhagic shock. Our aim was to study the potential role of nitric oxide synthases (NOS) in the cardiovascular response in hemorrhagic hypotension produced experimentally in anesthetized rats. METHODS: Groups of animals (n = 14, per group): (a) normotensive; (b) hypovolemic (20% blood loss); (c) normotensive and pretreatment with N(G)-nitro-L-arginine methyl ester (L-NAME); (d) hypovolemic and pretreatment with L-NAME. RESULTS: L-NAME restored the hypotension induced by hemorrhage. Blood loss decreased heart rate in the first stage increasing at 60 and 120 min. L-NAME blunted this effect. Right atria and left ventricle histochemical NOS activities increased at 60 and 120 min (atria 8% and 24%, respectively; ventricle 21% and 45%, respectively). This activity increased 17% in smooth muscle at 120 min. Heart endothelial NOS protein levels increased in heart at 60 min being attenuated at 120 min. Inducible NOS protein levels raised significantly in right atria, left ventricle and aorta at 120 min. CONCLUSION: Hemorrhagic shock induced a time-dependent and specific NOS activation modulating cardiovascular function. The inhibition of nitric oxide system appears to prevent the acceleration of heart rate during late phases after acute hypovolemic state induced by blood loss.


Asunto(s)
Aorta Torácica/enzimología , Hemorragia/enzimología , Hipovolemia/enzimología , Miocardio/enzimología , Óxido Nítrico Sintasa de Tipo III/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Animales , Aorta Torácica/efectos de los fármacos , Aorta Torácica/patología , Presión Sanguínea , Inhibidores Enzimáticos/farmacología , Corazón/efectos de los fármacos , Frecuencia Cardíaca , Hemodinámica , Hemorragia/patología , Hemorragia/fisiopatología , Hipovolemia/patología , Hipovolemia/fisiopatología , Masculino , Miocardio/patología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa de Tipo II/antagonistas & inhibidores , Óxido Nítrico Sintasa de Tipo III/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Factores de Tiempo
17.
J Endocrinol ; 230(2): 185-95, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270898

RESUMEN

This study aimed to investigate whether nitric oxide participates in the cardiovascular function and haemodynamic adaptation to acute haemorrhage in animals with thyroid disorders. Sprague-Dawley rats aged 2months old treated with T3 (hyper, 20µg/100g body weight) or 0.02% methimazole (hypo, w/v) during 28days were pre-treated with N(G) nitro-l-arginine methyl ester (L-NAME) and submitted to 20% blood loss. Heart function was evaluated by echocardiography. Measurements of arterial blood pressure, heart rate, nitric oxide synthase activity and protein levels were performed. We found that hypo decreased fractional shortening and ejection fraction and increased left ventricle internal diameter. Hyper decreased ventricle diameter and no changes in cardiac contractility. Haemorrhage elicited a hypotension of similar magnitude within 10min. Then, this parameter was stabilized at about 30-40min and maintained until finalized, 120min. L-NAME rats showed that the immediate hypotension would be independent of nitric oxide. Nitric oxide synthase inhibition blunted the changes of heart rate induced by blood loss. Hyper and hypo had lower atrial enzyme activity associated with a decreased enzyme isoform in hypo. In ventricle, hyper and hypo had a higher enzyme activity, which was not correlated with changes in protein levels. Haemorrhage induced an increased heart nitric oxide production. We concluded that thyroid disorders were associated with hypertrophic remodelling which impacted differently on cardiac function and its adaptation to a hypovolemia. Hypovolemia triggered a nitric oxide synthase activation modulating the heart function to maintain haemodynamic homeostasis. This involvement depends on a specific enzyme isoform, cardiac chamber and thyroid state.


Asunto(s)
Enfermedades Cardiovasculares/etiología , Óxido Nítrico/metabolismo , Enfermedades de la Tiroides/complicaciones , Adaptación Fisiológica , Animales , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Ecocardiografía , Hemodinámica , Hemorragia/fisiopatología , Hipovolemia/fisiopatología , Masculino , Distribución Aleatoria , Ratas Sprague-Dawley , Enfermedades de la Tiroides/diagnóstico por imagen , Enfermedades de la Tiroides/metabolismo , Enfermedades de la Tiroides/fisiopatología , Hormonas Tiroideas/deficiencia
18.
Biomed Pharmacother ; 81: 225-234, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27261598

RESUMEN

Aquaporin-1 (AQP1) is expressed in the heart and its relationship with NO system has not been fully explored. The aims of this work were to study the effects of NO system inhibition on AQP1 abundance and localization and evaluate AQP1 S-nitrosylation in a model of water restriction during postnatal growth. Rats aged 25 and 50days (n=15) were divided in: R: water restriction; C: water ad libitum; RL: L-NAME (4mg/kgday)+water restriction; CL: L-NAME+water ad libitum. AQP1 protein levels, immunohistochemistry and S-nitrosylation (colocalization of AQP1 and S-nitrosylated cysteines by confocal microscopy) were determined in cardiac tissue. We also evaluated the effects of NO donor sodium nitroprusside (SNP) on osmotic water permeability of cardiac membrane vesicles by stopped-flow spectrometry. AQP1 was present in cardiac vascular endothelium and endocardium in C and CL animals of both ages. Cardiac AQP1 levels were increased in R50 and RL50 and appeared in cardiomyocyte plasma membrane. No changes in AQP1 abundance or localization were observed in R25, but RL25 group showed AQP1 presence on cardiomyocyte sarcolemma. AQP1 S-nitrosylation was increased in R25 group, without changes in the 50-day-old group. Cardiac membrane vesicles expressing AQP1 presented a high water permeability coefficient and pretreatment with SNP decreased water transport. Age-related influence of NO system on AQP1 abundance and localization in the heart may affect cardiac water homeostasis during hypovolemic state. Increased AQP1 S-nitrosylation in the youngest group may decrease osmotic water permeability of cardiac membranes, having a negative impact on cardiac water balance.


Asunto(s)
Acuaporina 1/metabolismo , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico/metabolismo , Presión Osmótica , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Membrana Celular/efectos de los fármacos , Membrana Celular/metabolismo , Permeabilidad de la Membrana Celular/efectos de los fármacos , Ventrículos Cardíacos/efectos de los fármacos , Ventrículos Cardíacos/metabolismo , Inmunohistoquímica , Masculino , Membranas/efectos de los fármacos , Modelos Biológicos , NG-Nitroarginina Metil Éster/farmacología , Donantes de Óxido Nítrico/farmacología , Óxido Nítrico Sintasa/metabolismo , Nitrosación , Presión Osmótica/efectos de los fármacos , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Dispersión de Radiación , Sístole/efectos de los fármacos , Agua
19.
Biomed Pharmacother ; 69: 380-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25661386

RESUMEN

Previous work done in our laboratory showed that water restriction during 24 and 72h induced changes in cardiovascular NOS activity without altering NOS protein levels in young and adult animals. These findings indicate that the involvement of NO in the regulatory mechanisms during dehydration depends on the magnitude of the water restriction and on age. Our aim was to study whether a controlled water restriction of 1 month affects cardiac function, NO synthase (NOS) activity and NOS, and cav-1 and -3 protein levels in rats during aging. Male Sprague-Dawley rats aged 2 and 16 months were divided into 2 groups: (CR) control restriction (WR) water restriction. Measurements of arterial blood pressure, heart rate, oxidative stress, NOS activity and NOS/cav-1 and -3 protein levels were performed. Cardiac function was evaluated by echocardiography. The results showed that adult rats have greater ESV, EDV and SV than young rats with similar SBP. Decreased atria NOS activity was caused by a reduction in NOS protein levels. Adult animals showed increased cav-1. Water restriction decreased NOS activity in young and adult rats associated to an increased cav-1. TBARS levels increased in adult animals. Higher ventricular NOS activity in adulthood would be caused by a reduction in both cav. Water restriction reduced NOS activity and increased cav in both age groups. In conclusion, our results indicated that dehydration modifies cardiac NO system activity and its regulatory proteins cav in order to maintain physiological cardiac function. Functional alterations are induced by the aging process as well as hypovolemic state.


Asunto(s)
Envejecimiento/metabolismo , Caveolinas/metabolismo , Miocardio/metabolismo , Miocardio/patología , Óxido Nítrico/metabolismo , Presión Osmótica , Animales , Presión Sanguínea , Peso Corporal , Electrocardiografía , Conducta Alimentaria , Fibrosis , Frecuencia Cardíaca , Ventrículos Cardíacos/metabolismo , Hemodinámica , Masculino , Miocardio/enzimología , Óxido Nítrico Sintasa/metabolismo , Ratas Sprague-Dawley , Sístole , Sustancias Reactivas al Ácido Tiobarbitúrico/metabolismo , Orina
20.
Nephron Physiol ; 95(2): p36-42, 2003.
Artículo en Inglés | MEDLINE | ID: mdl-14610322

RESUMEN

BACKGROUND: Reduction in renal mass is associated with several structural and functional adaptations including compensatory renal growth and hemodynamic changes. The mediators of the renal hemodynamic adaptations have not been definitively identified. Several investigators have postulated that nitric oxide (NO) is involved this physiological mechanisms. The purpose of this study was to evaluate the role of vascular and renal NO pathway in the model of subtotal nephrectomy-salt load hypertension. MATERIALS AND METHODS: Wistar rats with 75% renal mass reduction (RMR) and saline load were studied during 4 weeks. Weekly, indirect systolic blood pressure (SBP) were measured. One week after nephrectomy, animals were divided in two groups, hypertensive (SBP > 140 mm Hg) and normotensive (SBP < 140 mm Hg). Urinary excretion of nitrates and nitrites (NOx), urinary chemioluminiscence levels and NOS activity in the left kidney and in the thoracic aorta artery were determined at the fourth week after subtotal nephrectomy. RESULTS: Urinary excretion of sodium was higher in normotensive rats than hypertensive rats and in both groups this parameter was higher than in sham rats. NOx excretion and NOS activity in the different nephron segments were higher in normotensive rats than in the hypertensive ones. In contrast, NOS activity in aorta sections and urinary chemiluminescence levels in hypertensive animals were enhanced compared with normotensive rats. These parameters were higher in both groups of nephrectomized rats than in sham ones. CONCLUSION: This study provides evidence to support the fact that the activation of the renal NO system is an important mechanism whereby the remnant kidney regulates sodium and water balance, contributing to control the arterial blood pressure in the renal mass reduction and saline load model.


Asunto(s)
Presión Sanguínea/fisiología , Hipertensión Renal/metabolismo , Hipertensión Renal/fisiopatología , Riñón/metabolismo , Riñón/fisiopatología , Óxido Nítrico/fisiología , Animales , Modelos Animales de Enfermedad , Tasa de Filtración Glomerular/fisiología , Hipertensión Renal/etiología , Riñón/cirugía , Masculino , Nefrectomía/métodos , Ratas , Ratas Wistar , Solución Salina Hipertónica/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA