Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Life Sci ; 82(15-16): 847-54, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18302965

RESUMEN

Atrial natriuretic peptide (ANP) exerts beneficial effects on the cardiovascular system in part by exerting antioxidant activity. Given that oxidant stress is a key cause of endothelial dysfunction in diabetes, we investigated whether ANP improves endothelial function in rats with diabetes. Rats were injected with streptozotocin (55 mg/kg iv) to induce type 1 diabetes or the citrate vehicle as controls (n=12). After 4 weeks the diabetic rats were treated with ANP (10 pmol/kg/min sc, n=12) or the antioxidant tempol (1.5 mmol/kg/day sc, n=11), both by osmotic minipump, ramipril (1 mg/kg per day in the drinking water) or remained untreated (n=11). After a further 4 weeks, anaesthetised rats were killed by exsanguination and the thoracic aortae collected for examination of vascular activity and measurement of superoxide generation. Diabetic rats showed elevated plasma glucose concentration (45+/-3 mM) compared to controls (10+/-1 mM) and this was not affected by ANP (43+/-3 mM), ramipril (41+/-2 mM) or tempol (43+/-2 mM). Endothelium-dependent relaxation ex vivo in response to acetylcholine was impaired in diabetic rats (Rmax=66+/-4%) compared to control rats (Rmax=94+/-1%) but treatment with ANP (Rmax=80+/-4%), ramipril (Rmax=88+/-2%) or tempol (Rmax=81+/-5%) significantly improved those responses. Relaxant responses to the endothelium-independent vasodilator sodium nitroprusside were enhanced by treatment of diabetic rats with ANP or ramipril and their combination; but not by tempol. Superoxide generation was significantly elevated in aorta from untreated diabetic rats (649+/-146% of control). In diabetic rats, superoxide generation was significantly attenuated by ANP (to 229+/-78%) or tempol (to 186+/-64%). This study demonstrates that ANP improves vascular oxidant stress in concert with endothelial function, independent of any effect on plasma glucose levels. These studies may lead to new therapies, based on natriuretic peptide and/or antioxidant approaches, for ameliorating the vascular complications of diabetes.


Asunto(s)
Factor Natriurético Atrial/farmacología , Angiopatías Diabéticas/prevención & control , Endotelio Vascular/patología , Envejecimiento/fisiología , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Animales , Antioxidantes/farmacología , Aorta Torácica/efectos de los fármacos , Aorta Torácica/metabolismo , Glucemia/metabolismo , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Western Blotting , Peso Corporal/efectos de los fármacos , Óxidos N-Cíclicos/farmacología , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Angiopatías Diabéticas/patología , Endotelio Vascular/metabolismo , Masculino , Relajación Muscular/efectos de los fármacos , NADPH Oxidasas/metabolismo , Óxido Nítrico Sintasa de Tipo III/biosíntesis , Nitroprusiato/farmacología , Ramipril/farmacología , Ratas , Ratas Sprague-Dawley , Marcadores de Spin , Superóxidos/metabolismo , Vasodilatadores/farmacología
2.
Antioxid Redox Signal ; 9(1): 101-13, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17115889

RESUMEN

The objective was a comprehensive investigation of the mechanisms and sites of resveratrol cardioprotection during and following ischemia-reperfusion (I-R) injury, and to determine whether direct preservation of cardiomyocytes is an important site of cardioprotection. We now provide the first definitive evidence that resveratrol specifically protects cardiomyocytes from I-R injury via a combination of suppression of superoxide levels and activation of potassium channels. This protection is apparent whether resveratrol is present for the full duration of the insult or only on recovery. In addition, resveratrol improved postischemic recovery of left ventricular contractile function, attenuated myocardial injury, and increased myocardial activation of the survival kinase Akt in the intact heart. Furthermore, resveratrol elicited direct concentration-dependent protective actions on the vasculature (vasorelaxation, superoxide suppression) and enhanced endothelium-dependent vasodilatation. Resveratrol thus targets a number of consequences of myocardial I-R, including release of reactive oxygen species, loss of recovery of contractile function, and impaired endothelium-dependent vasodilatation. Previous evidence indicates that resveratrol elicits potent preconditioning in the heart. Given that myocardial ischemic events are often unpredictable in humans, the findings that resveratrol protection is also evident when administered during and/or after the insult adds new dimensions to the clinical potential of resveratrol.


Asunto(s)
Antioxidantes/farmacología , Corazón/efectos de los fármacos , Miocitos Cardíacos/efectos de los fármacos , Daño por Reperfusión/prevención & control , Estilbenos/farmacología , Animales , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Humanos , Hipoxia , Técnicas In Vitro , Masculino , Ratas , Ratas Sprague-Dawley , Resveratrol
3.
Cardiovasc Res ; 72(1): 112-23, 2006 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-16890211

RESUMEN

OBJECTIVE: Reactive oxygen species (ROS) such as superoxide have been linked to the hypertrophic response of the heart to stimuli including angiotensin II (AngII), mechanical stretch, and pressure overload. We have previously demonstrated that cGMP and protein kinase G mediate the antihypertrophic actions of the natriuretic peptides in rat cardiomyocytes and isolated whole hearts. The impact of natriuretic peptides on cardiac ROS generation, however, has not been investigated. We tested the hypothesis that reduced superoxide accumulation contributes to the antihypertrophic action of atrial natriuretic peptide (ANP). METHODS: Neonatal rat cardiomyocytes were cultured in serum-free medium with and without AngII (1 micromol/L) or endothelin-1 (ET(1), 60 nmol/L) in the presence and absence of ANP (1 micromol/L) or tempol (100 micromol/L). Hypertrophic responses, cardiomyocyte superoxide generation, and cardiomyocyte expression of NADPH oxidase were determined. RESULTS: AngII induced increases in cardiomyocyte size (to 176 +/- 9% n = 8 p < 0.001, at 48 h), beta-myosin heavy chain expression (to 4.0 +/- 1.6-fold n = 6 p < 0.05, at 48 h), c-fos expression (to 1.9 +/- 0.5-fold n = 7 p < 0.01, at 6 h), superoxide generation (to 181+/-21% n = 8 p < 0.005, at 24 h), and expression of the gp91phox subunit of NADPH oxidase (to 2.4 +/- 0.5-fold n = 7 p < 0.05, at 48 h). These effects were all significantly inhibited by ANP: cardiomyocyte size, beta-myosin heavy chain expression, c-fos expression, superoxide generation and gp91phox expression were reduced to 107 +/- 5% (n = 5 p < 0.05), 1.2 +/- 0.2-fold (n = 6 p < 0.05), 0.9 +/- 0.2-fold (n = 7 p < 0.05), 141 +/- 21% (n = 8 p < 0.05), and to 1.0 +/- 0.5-fold (n = 7 p < 0.05), respectively. These effects were mimicked by tempol. ANP and tempol also significantly inhibited ET1-induced increases in cardiomyocyte size and superoxide generation, but had no effect on markers of hypertrophy when studied alone. CONCLUSION: This data indicates that the antihypertrophic actions of ANP are accompanied by reduced levels of superoxide, suggesting an antioxidant action contributes to the antihypertrophic actions of ANP.


Asunto(s)
Antioxidantes/farmacología , Factor Natriurético Atrial/farmacología , Miocitos Cardíacos/metabolismo , Angiotensina II/farmacología , Animales , Animales Recién Nacidos , Aumento de la Célula/efectos de los fármacos , Células Cultivadas , Óxidos N-Cíclicos/farmacología , Endotelina-1/farmacología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Genes fos , Miocitos Cardíacos/efectos de los fármacos , Cadenas Pesadas de Miosina/genética , NADPH Oxidasas/genética , Ratas , Ratas Sprague-Dawley , Marcadores de Spin , Superóxidos/análisis
4.
Diab Vasc Dis Res ; 14(5): 423-433, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28565941

RESUMEN

Diabetic cardiomyopathy is a major contributor to the increasing burden of heart failure globally. Effective therapies remain elusive, in part due to the incomplete understanding of the mechanisms underlying diabetes-induced myocardial injury. The objective of this study was to assess the direct impact of insulin replacement on left ventricle structure and function in a rat model of diabetes. Male Sprague-Dawley rats were administered streptozotocin (55 mg/kg i.v.) or citrate vehicle and were followed for 8 weeks. A subset of diabetic rats were allocated to insulin replacement (6 IU/day insulin s.c.) for the final 4 weeks of the 8-week time period. Diabetes induced the characteristic systemic complications of diabetes (hyperglycaemia, polyuria, kidney hypertrophy) and was accompanied by marked left ventricle remodelling (cardiomyocyte hypertrophy, left ventricle collagen content) and diastolic dysfunction (transmitral E/A, left ventricle-dP/dt). Importantly, these systemic and cardiac impairments were ameliorated markedly following insulin replacement, and moreover, markers of the diabetic cardiomyopathy phenotype were significantly correlated with the extent of hyperglycaemia. In summary, these data suggest that poor glucose control directly contributes towards the underlying features of experimental diabetic cardiomyopathy, at least in the early stages, and that adequate replacement ameliorates this.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Cardiomiopatías Diabéticas/prevención & control , Ventrículos Cardíacos/efectos de los fármacos , Hipoglucemiantes/farmacología , Insulina/farmacología , Estreptozocina , Función Ventricular Izquierda/efectos de los fármacos , Animales , Biomarcadores/sangre , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inducido químicamente , Cardiomiopatías Diabéticas/etiología , Cardiomiopatías Diabéticas/patología , Cardiomiopatías Diabéticas/fisiopatología , Progresión de la Enfermedad , Fibrosis , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Masculino , Miocarditis/patología , Miocarditis/fisiopatología , Miocarditis/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Estrés Oxidativo/efectos de los fármacos , Ratas Sprague-Dawley , Factores de Tiempo , Remodelación Ventricular/efectos de los fármacos
5.
Eur J Pharmacol ; 807: 12-20, 2017 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-28438648

RESUMEN

Endoplasmic reticulum (ER) stress contributes to progression of diabetic nephropathy, which promotes end-stage renal failure in diabetic patients. This study was undertaken to investigate the actions of tempol and ramipril, pharmacological agents that target the consequences of NADPH oxidase, on diabetic nephropathy in a rat model of type 1 diabetes, with an emphasis on markers of ER stress. Male Sprague-Dawley rats were injected intravenously with a single bolus of streptozotocin (55mg/kg) to induce type 1 diabetes. An additional age-matched group of rats was administered with citrate vehicle as controls. After 4 weeks of untreated diabetes, rats received tempol (1.5mM/kg/day subcutaneously, n=8), ramipril (1mg/kg/day in drinking water, n=8) or remained untreated for an additional 4 weeks (n=7). After 8 weeks of diabetes in total, kidneys were collected for histological analysis, gene expression and protein abundance. Tempol and ramipril blunted diabetes-induced upregulation of NADPH oxidase isoforms (Nox4, Nox2, p47phox), accompanied by an amelioration of diabetes-induced glomerular injury (podocin, nephrin, Kim-1), tubulo-interstitial fibrosis (TGFß1, TGFß-R2, pSMAD3, α-SMA) and pro-inflammatory cytokines (TNFα, MCP-1, ANX-A1, FPR2) expression. In addition, the diabetes-induced renal ER stress, evidenced by increased expression of GRP-78 chaperone and stress-associated markers ATF4, TRB3, as well as XBP1s, phospho-p38 mitogen-activated protein kinase (MAPK) and 3-nitrotyrosination, were all attenuated by tempol and ramipril. These observations suggest that antioxidant approaches that blunt NADPH upregulation may attenuate diabetic nephropathy, at least in part by negatively regulating ER stress and inflammation, and hence ameliorating kidney damage.


Asunto(s)
Materiales Biomiméticos/farmacología , Óxidos N-Cíclicos/farmacología , Nefropatías Diabéticas/tratamiento farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , NADPH Oxidasas/metabolismo , Superóxido Dismutasa/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Animales , Materiales Biomiméticos/uso terapéutico , Óxidos N-Cíclicos/uso terapéutico , Nefropatías Diabéticas/metabolismo , Nefropatías Diabéticas/patología , Modelos Animales de Enfermedad , Fibrosis , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Túbulos Renales/efectos de los fármacos , Túbulos Renales/patología , Masculino , Óxido Nítrico/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ramipril/farmacología , Ratas , Ratas Sprague-Dawley , Marcadores de Spin
6.
Br J Pharmacol ; 145(4): 495-502, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15821756

RESUMEN

1 The anti-inflammatory properties of annexin-1 peptides have been largely ascribed to their powerful antineutrophil actions in vivo. We have recently reported that the N-terminal fragment of annexin-1, Anx-1(2-26), preserves contractile function of cardiac muscle in vitro. The aim of the present study was to determine if Anx-1(2-26) elicits protective actions specifically on the cardiac myocyte (in the absence of neutrophils), using a model of metabolic inhibition to simulate ischaemia. 2 Metabolic inhibition of cardiac myocytes (4 h incubation at 37 degrees C in HEPES-containing buffer supplemented with 2-deoxy-D-glucose, D,L-lactic acid and pH adjusted to 6.5) followed by 2.5 h recovery in normal medium markedly increased creatine kinase (CK) and lactate dehydrogenase (LDH) levels by 179+/-39 and 26+/-7 IU L(-1) (both n=40, P<0.001), respectively. However, cellular injury was significantly decreased when Anx-1(2-26) (0.3 microM) was present during metabolic inhibition, CK by 74+/-10% and LDH by 71+/-6% (both n=31, P<0.001), respectively. 3 Boc 2 (10 microM), a nonselective formyl peptide receptor antagonist, present during metabolic inhibition, abolished the cardioprotective effect of Anx-1(2-26). 4 Addition of chelerythrine (10 microM), 5-hydroxydecanoate (500 microM) or SB202190 (1 microM) during metabolic inhibition also abolished Anx-1(2-26)-induced cardioprotection. 5 Cellular injury induced by metabolic inhibition was also largely prevented when myocytes were incubated with Anx-1(2-26) for 5 min with 10 min recovery prior to the insult, or when Anx-1(2-26) was present only during the recovery period following drug-free metabolic inhibition. 6 In conclusion, the annexin-1 peptide Anx-1(2-26) potently prevents cardiac myocyte injury induced by metabolic inhibition, an action that was dependent at least in part on the activation of the formyl peptide receptor family of G-protein-coupled receptors, protein kinase C, p38 mitogen-activated protein kinase and ATP-sensitive potassium channels.


Asunto(s)
Anexina A1/farmacología , Miocitos Cardíacos/efectos de los fármacos , Alcaloides , Análisis de Varianza , Animales , Benzofenantridinas , Células Cultivadas , Creatina Quinasa/metabolismo , Ácidos Decanoicos/farmacología , Inhibidores Enzimáticos/farmacología , Hidroxiácidos/farmacología , Imidazoles/farmacología , L-Lactato Deshidrogenasa/metabolismo , Masculino , Isquemia Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Péptidos , Fenantridinas/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
7.
Br J Pharmacol ; 168(1): 238-52, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22924634

RESUMEN

BACKGROUND AND PURPOSE: Annexin-A1 (ANX-A1) is an endogenous, glucocorticoid-regulated anti-inflammatory protein. The N-terminal-derived peptide Ac-ANX-A1(2-26) preserves cardiomyocyte viability, but the impact of ANX-A1-peptides on cardiac contractility is unknown. We now test the hypothesis that ANX-A1 preserves post-ischaemic recovery of left ventricular (LV) function. EXPERIMENTAL APPROACH: Ac-ANX-A1(2-26) was administered on reperfusion, to adult rat cardiomyocytes as well as hearts isolated from rats, wild-type mice and mice deficient in endogenous ANX-A1 (ANX-A1(-/-)). Myocardial viability and recovery of LV function were determined. KEY RESULTS: Ischaemia-reperfusion markedly impaired both cardiomyocyte viability and recovery of LV function by 60%. Treatment with exogenous Ac-ANX-A1(2-26) at the onset of reperfusion prevented cardiomyocyte injury and significantly improved recovery of LV function, in both intact rat and wild-type mouse hearts. Ac-ANX-A1(2-26) cardioprotection was abolished by either formyl peptide receptor (FPR)-nonselective or FPR1-selective antagonists, Boc2 and cyclosporin H, but was relatively insensitive to the FPR2-selective antagonist QuinC7. ANX-A1-induced cardioprotection was associated with increased phosphorylation of the cell survival kinase Akt. ANX-A1(-/-) exaggerated impairment of post-ischaemic recovery of LV function, in addition to selective LV FPR1 down-regulation. CONCLUSIONS AND IMPLICATIONS: These data represent the first evidence that ANX-A1 affects myocardial function. Our findings suggest ANX-A1 is an endogenous regulator of post-ischaemic recovery of LV function. Furthermore, the ANX-A1-derived peptide Ac-ANX-A1(2-26) on reperfusion rescues LV function, probably via activation of FPR1. ANX-A1-based therapies may thus represent a novel clinical approach for the prevention and treatment of myocardial reperfusion injury.


Asunto(s)
Anexina A1/metabolismo , Cardiotónicos/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/metabolismo , Fragmentos de Péptidos/farmacología , Disfunción Ventricular Izquierda/prevención & control , Animales , Anexina A1/deficiencia , Anexina A1/farmacología , Regulación hacia Abajo , Femenino , Técnicas In Vitro , Masculino , Ratones , Contracción Miocárdica , Daño por Reperfusión Miocárdica/complicaciones , Fosforilación , Ratas , Ratas Sprague-Dawley , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/metabolismo
8.
PLoS One ; 7(4): e34892, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22506056

RESUMEN

BACKGROUND: New therapeutic targets for cardiac hypertrophy, an independent risk factor for heart failure and death, are essential. HNO is a novel redox sibling of NO• attracting considerable attention for the treatment of cardiovascular disorders, eliciting cGMP-dependent vasodilatation yet cGMP-independent positive inotropy. The impact of HNO on cardiac hypertrophy (which is negatively regulated by cGMP) however has not been investigated. METHODS: Neonatal rat cardiomyocytes were incubated with angiotensin II (Ang II) in the presence and absence of the HNO donor Angeli's salt (sodium trioxodinitrate) or B-type natriuretic peptide, BNP (all 1 µmol/L). Hypertrophic responses and its triggers, as well as cGMP signaling, were determined. RESULTS: We now demonstrate that Angeli's salt inhibits Ang II-induced hypertrophic responses in cardiomyocytes, including increases in cardiomyocyte size, de novo protein synthesis and ß-myosin heavy chain expression. Angeli's salt also suppresses Ang II induction of key triggers of the cardiomyocyte hypertrophic response, including NADPH oxidase (on both Nox2 expression and superoxide generation), as well as p38 mitogen-activated protein kinase (p38MAPK). The antihypertrophic, superoxide-suppressing and cGMP-elevating effects of Angeli's salt were mimicked by BNP. We also demonstrate that the effects of Angeli's salt are specifically mediated by HNO (with no role for NO• or nitrite), with subsequent activation of cardiomyocyte soluble guanylyl cyclase (sGC) and cGMP signaling (on both cGMP-dependent protein kinase, cGK-I and phosphorylation of vasodilator-stimulated phosphoprotein, VASP). CONCLUSIONS: Our results demonstrate that HNO prevents cardiomyocyte hypertrophy, and that cGMP-dependent NADPH oxidase suppression contributes to these antihypertrophic actions. HNO donors may thus represent innovative pharmacotherapy for cardiac hypertrophy.


Asunto(s)
Cardiomegalia/tratamiento farmacológico , Guanilato Ciclasa/metabolismo , Miocitos Cardíacos/efectos de los fármacos , Óxidos de Nitrógeno/farmacología , Receptores Citoplasmáticos y Nucleares/metabolismo , Superóxidos/metabolismo , Angiotensina II/efectos adversos , Animales , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Cardiomegalia/patología , Moléculas de Adhesión Celular/metabolismo , GMP Cíclico/metabolismo , Endotelina-1/metabolismo , Proteínas de Microfilamentos/metabolismo , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , NADPH Oxidasas/metabolismo , Péptido Natriurético Encefálico/metabolismo , Nitritos/farmacología , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Especies Reactivas de Oxígeno/antagonistas & inhibidores , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Guanilil Ciclasa Soluble , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
J Cardiovasc Pharmacol ; 47(1): 1-8, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16424779

RESUMEN

The cardioprotective actions of nitric oxide (NO) have largely been attributed to cGMP. NO may, however, elicit some biological actions independently of cGMP. We tested the hypothesis that the NO donor sodium nitroprusside specifically protects isolated cardiomyocytes from injury at least in part independently of its ability to elevate cGMP by using metabolic inhibition to simulate ischemia. Metabolic inhibition-induced injury of adult rat cardiomyocytes (increased activity of lactate dehydrogenase and creatine kinase) was significantly reduced by sodium nitroprusside by at least 30% at all concentrations studied (0.3-100 microM). Sodium nitroprusside (1 microM) increased cardiomyocyte cGMP content, but neither a stable analogue of cGMP (8-bromo-cGMP) nor a potent cGMP stimulus (atrial natriuretic peptide) mimicked the protective effects of sodium nitroprusside. Moreover, inhibition of soluble guanylyl cyclase failed to inhibit sodium nitroprusside cardiomyocyte protection. Conversely, inhibition of either ATP-sensitive potassium (K(ATP)) channels with glibenclamide (10 microM) or calcium-sensitive potassium (K(Ca)) channels with tetraethylammonium bromide (1 mM) or iberiotoxin (20 nM) markedly attenuated the cardioprotective actions of sodium nitroprusside. In conclusion, sodium nitroprusside protects isolated cardiomyocytes from metabolic inhibition independently of cGMP; rather, inhibition of K(Ca) and K(ATP) channels reverses the sodium nitroprusside actions, thus unmasking another mechanism for NO-mediated protection in cardiomyocytes.


Asunto(s)
GMP Cíclico/fisiología , Isquemia Miocárdica/patología , Daño por Reperfusión Miocárdica/prevención & control , Miocitos Cardíacos/efectos de los fármacos , Óxido Nítrico/fisiología , Nitroprusiato/farmacología , Animales , Masculino , Miocitos Cardíacos/patología , Canales de Potasio/fisiología , Ratas , Ratas Sprague-Dawley
10.
Phytother Res ; 19(11): 932-7, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16317648

RESUMEN

This study aims to investigate the possible hepato-protective effects of honokiol against liver damage and cirrhosis induced by carbon tetrachloride (CCl(4)) in the rat. Rats were treated acutely, or chronically with CCl(4) at 5 day intervals (0.06 mL/100 g body weight, administered as 50% vol/vol solution in liquid paraffin) by gavage, in combination with phenobarbitone in drinking water (0.5 g/L for 7 days prior to, and during CCl(4) treatment) to induce liver damage. Some were also co-treated with 0.1 mg/kg or 0.03 mg/kg honokiol (i.p.) or with appropriate vehicle. In vivo measurement of the liver sinusoidal area was performed using confocal microscopy following i.v. fluorescein isothiocyanate (FITC) dextran. Liver histology and function tests were performed, and liver and body weights were measured. Confocal microscopy showed that acute and chronic CCl(4) treatment significantly reduced the sinusoidal area. Honokiol (0.1 mg/kg, but not 0.03 mg/kg) partially reversed the decrease in the sinusoidal area after acute or chronic treatments with CCl(4). Acute and chronic CCl(4) treatment produced significant histological liver damage. Honokiol (0.1 mg/kg) significantly reduced the histological damage caused by chronic treatment. Chronic treatment with CCl(4) caused a significant increase in the bilirubin level that was not observed following the high dose of honokiol (0.1 mg/kg). In conclusion, this study showed that honokiol exhibits potent hepato-protective effects in rats treated with CCl(4).


Asunto(s)
Compuestos de Bifenilo/uso terapéutico , Lignanos/uso terapéutico , Cirrosis Hepática Experimental/tratamiento farmacológico , Animales , Bilirrubina/sangre , Tetracloruro de Carbono , Enfermedad Veno-Oclusiva Hepática/prevención & control , Hígado/patología , Hígado/fisiopatología , Cirrosis Hepática Experimental/patología , Cirrosis Hepática Experimental/fisiopatología , Magnolia , Masculino , Microscopía Confocal , Ratas , Ratas Long-Evans
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda