Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
J Cell Mol Med ; 23(7): 4779-4794, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31087547

RESUMEN

The thiazolidinedione (TZD) class of Peroxisome proliferator-activated receptor gamma agonists has restricted clinical use for diabetes mellitus due to fluid retention and potential cardiovascular risks. These side effects are attributed in part to direct salt-retaining effect of TZDs at the renal collecting duct. A recent study from our group revealed that prolonged rosiglitazone (RGZ) treatment caused no Na+/H2 O retention or up-regulation of Na+ transport-linked channels/transporters in experimental congestive heart failure (CHF) induced by surgical aorto-caval fistula (ACF). The present study examines the effects of RGZ on renal and cardiac responses to atrial natriuretic peptide (ANP), Acetylcholine (Ach) and S-Nitroso-N-acetylpenicillamine (SNAP-NO donor). Furthermore, we assessed the impact of RGZ on gene expression related to the ANP signalling pathway in animals with ACF. Rats subjected to ACF (or sham) were treated with either RGZ (30 mg/kg/day) or vehicle for 4 weeks. Cardiac chambers pressures and volumes were assessed invasively via Miller catheter. Kidney excretory and renal hemodynamic in response to ANP, Ach and SNAP were examined. Renal clearance along with cyclic guanosine monophosphate (cGMP), gene expression of renal CHF-related genes and ANP signalling in the kidney were determined. RGZ-treated CHF rats exhibited significant improvement in the natriuretic responses to ANP infusion. This 'sensitization' to ANP was not associated with increases in neither urinary cGMP nor in vitro cGMP production. However, RGZ caused down-regulation of several genes in the renal cortex (Ace, Nos3 and Npr1) and up-regulation of ACE2, Agtrla, Mme and Cftr along down-regulation of Avpr2, Npr1,2, Nos3 and Pde3 in the medulla. In conclusion, CHF+RGZ rats exhibited significant enhancement in the natriuretic responses to ANP infusion, which are known to be blunted in CHF. This 'sensitization' to ANP is independent of cGMP signalling, yet may involve post-cGMP signalling target genes such as ACE2, CFTR and V2 receptor. The possibility that TZD treatment in uncomplicated CHF may be less detrimental than thought before deserves additional investigations.


Asunto(s)
Factor Natriurético Atrial/uso terapéutico , Insuficiencia Cardíaca/tratamiento farmacológico , Riñón/patología , Rosiglitazona/uso terapéutico , Acetilcolina/farmacología , Animales , Factor Natriurético Atrial/administración & dosificación , Presión Sanguínea/efectos de los fármacos , GMP Cíclico/metabolismo , Endotelio/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/patología , Hemodinámica/efectos de los fármacos , Riñón/efectos de los fármacos , Masculino , Ratas Sprague-Dawley , Rosiglitazona/farmacología , Transducción de Señal/efectos de los fármacos , Vasodilatación/efectos de los fármacos
2.
Am J Nephrol ; 38(4): 292-9, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24080549

RESUMEN

BACKGROUND: The natriuretic/diuretic response to atrial natriuretic peptide (ANP), an important regulator of water and Na(+) balance, is markedly attenuated in nephrotic syndrome (NS). It has been suggested that the diminished renal responsiveness to ANP may contribute to the pathogenesis of salt retention and edema formation in NS. However, the mechanisms underlying the renal hyporesponsiveness to ANP remain largely unknown. METHODS: The acute effects of exogenous infusion of ANP (5 µg/kg + 10 µg/kg/h) were studied by clearance methodology in control rats, hypoalbuminemic rats with Adriamycin (ADR)-induced NS and in ADR-treated rats infused with hyperoncotic albumin sufficient to restore plasma albumin to normal levels. RESULTS: Administration of ANP to control rats resulted in a significant increase in urinary flow rate, absolute rate of sodium excretion (+456%) and glomerular filtration rate (GFR). Mean arterial blood pressure decreased following infusion of the peptide. In the nephrotic rats, baseline GFR and Na(+) excretion were significantly lower than in the control animals, and the renal effects of ANP were markedly blunt compared to the control animals. In contrast, the hypotensive effect of ANP in the ADR-treated rats was largely preserved. Infusion of hyperoncotic albumin prior to ANP administration reversed the decrease in baseline GFR and Na(+) excretion and completely restored the renal effects of ANP in the nephrotic rats. CONCLUSION: These findings indicate that renal hyporesponsiveness to ANP in rats with ADR-induced NS is a reversible phenomenon that appears to be of functional origin rather than reflecting permanent cellular damage.


Asunto(s)
Albúminas/farmacología , Factor Natriurético Atrial/metabolismo , Riñón/efectos de los fármacos , Síndrome Nefrótico/tratamiento farmacológico , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Edema/inducido químicamente , Tasa de Filtración Glomerular , Humanos , Hipotensión/metabolismo , Inulina/farmacocinética , Masculino , Natriuresis/efectos de los fármacos , Nefrosis/patología , Ratas , Ratas Sprague-Dawley , Circulación Renal/efectos de los fármacos , Renina/sangre , Sales (Química) , Sodio/metabolismo
3.
J Biomed Biotechnol ; 2011: 729497, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21274403

RESUMEN

Despite continuous progress in our understanding of the pathogenesis of congestive heart failure (CHF) and its management, mortality remains high. Therefore, development of reliable experimental models of CHF and cardiac hypertrophy is essential to better understand disease progression and allow new therapy development. The aortocaval fistula (ACF) model, first described in dogs almost a century ago, has been adopted in rodents by several groups including ours. Although considered to be a model of high-output heart failure, its long-term renal and cardiac manifestations are similar to those seen in patients with low-output CHF. These include Na+-retention, cardiac hypertrophy and increased activity of both vasoconstrictor/antinatriureticneurohormonal systems and compensatory vasodilating/natriuretic systems. Previous data from our group and others suggest that progression of cardiorenal pathophysiology in this model is largely determined by balance between opposing hormonal forces, as reflected in states of CHF decompensation that are characterized by overactivation of vasoconstrictive/Na+-retaining systems. Thus, ACF serves as a simple, cheap, and reproducible platform to investigate the pathogenesis of CHF and to examine efficacy of new therapeutic approaches. Hereby, we will focus on the neurohormonal, renal, and cardiac manifestations of the ACF model in rats, with special emphasis on our own experience.


Asunto(s)
Cardiomegalia/patología , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/patología , Fístula Vascular/patología , Animales , Ratas , Remodelación Ventricular
4.
Clin Exp Hypertens ; 33(7): 444-54, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21932990

RESUMEN

The Cohen-Rosenthal Diabetic Hypertensive rat (CRDH) is a unique animal model in which genetic hypertension and diabetes developed after crossbreeding of Cohen diabetic rats sensitive substrain (CDR) and spontaneously hypertensive rats (SHR). The present study examined: 1) The acute effects of ET-1 on the systemic and renal hemodynamics in CRDH rats, CDR, and SHR; 2) The expression of ET-1 and its receptors in the renal tissue of CRDH rats. Intravenous injection of ET-1 (1.0 nmol/kg) into anesthetized SHR rats resulted in a significant immediate depressor response (mean arterial pressure (MAP) decreased from 165 ± 3 to 124 ± 12 mmHg, p < 0.0001) followed by a minor hypertensive phase (MAP increased to 170 ± 2 mmHg). Simultaneously, the administration of ET-1 caused a significant decrease in renal blood flow (RBF) from 5.8 ± 0.9 ml/min to 3.2 ± 0.5 ml/min (p = 0.026). These responses were blunted in CRDH rats and CDR. Analysis of intra-renal blood flow by laser-Doppler in CRDH rats revealed that ET-1 injection caused a decrease in cortical blood flow (Δ = -12 ± 2.9%). However, in contrast to its well-known renal medullary vasodilatory effect, ET-1 produced a significant decline in the medulla blood flow (Δ = -17.5 ± 3.4%) (p = 0.0125). These findings suggest that CDR and CRDH rats have reduced sensitivity to vascular and renal action of ET-1. Furthermore, in the CRDH rats, the expected ET-1-induced medullary vasodilatation was abolished and even reversed into prolonged vasoconstriction.


Asunto(s)
Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/fisiopatología , Endotelina-1/farmacología , Endotelina-1/fisiología , Hemodinámica/fisiología , Hipertensión/complicaciones , Hipertensión/fisiopatología , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología , Animales , Secuencia de Bases , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Cartilla de ADN/genética , Diabetes Mellitus Experimental/genética , Antagonistas de los Receptores de la Endotelina A , Antagonistas de los Receptores de la Endotelina B , Endotelina-1/genética , Expresión Génica , Hemodinámica/efectos de los fármacos , Hipertensión/genética , Riñón/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas SHR , Ratas Endogámicas , Receptor de Endotelina A/genética , Receptor de Endotelina B/genética
5.
J Pharmacol Exp Ther ; 334(3): 820-9, 2010 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-20519551

RESUMEN

Peroxisome proliferator-activated receptor (PPAR)-gamma modulators, a class of antidiabetic drugs, have been associated with cardiovascular risks in type 2 diabetes in humans. The objective of this study was to explore possible cardiovascular risk biomarkers associated with PPAR-gamma in rodents that could provide an alert for risk to humans. Normal, myocardial infarction-induced heart failure (HF) or Zucker diabetic fatty (ZDF) rats were used. Rats (n = 5-6) were treated with either vehicle or rosiglitazone (RGZ; 3 or 45 mg/kg/day p.o.) for 4 weeks. Biomarkers for potential cardiovascular risks were assessed, including 1) ultrasound for cardiac structure and function; 2) neuroendocrine and hormonal plasma biomarkers of cardiovascular risk; 3) pharmacogenomic profiling of cardiac and renal tissue by targeted tissue low-density gene array representing ion channels and transporters, and components of the renin-angiotensin-aldosterone system; and 4) immunohistochemistry for cardiac fibrosis, hypertrophy, and inflammation (macrophages and tumor necrosis factor-alpha). HF was confirmed by increase in cardiac brain natriuretic peptide expression (p < 0.01) and echocardiography. Adequate exposure of RGZ was confirmed by pharmacokinetics (plasma drug levels) and the pharmacodynamic biomarker adiponectin. In normal or HF rats, RGZ had no negative effects on any of the biomarkers investigated. Similarly, RGZ had no significant effects on gene expression except for the increase in interleukin-6 mRNA expression in the heart and decrease in epithelial sodium channel beta in the kidney. In contrast, echocardiography showed improved cardiac structure and function after RGZ in ZDF rats. Taken together, this study suggests a limited predictive power of these preclinical models in respect to observed clinical adverse effects associated with RGZ.


Asunto(s)
Biomarcadores/sangre , Insuficiencia Cardíaca/inducido químicamente , Hipoglucemiantes/efectos adversos , PPAR gamma/agonistas , Tiazolidinedionas/efectos adversos , Animales , Peso Corporal , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Ecocardiografía , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/patología , Hemodinámica/fisiología , Hipoglucemiantes/farmacocinética , Inmunohistoquímica , Miocarditis/inducido químicamente , Miocarditis/patología , Miocardio/patología , Tamaño de los Órganos , ARN/genética , Ratas , Ratas Endogámicas Lew , Ratas Zucker , Rosiglitazona , Tiazolidinedionas/farmacocinética , Investigación Biomédica Traslacional
6.
Surg Endosc ; 24(4): 826-33, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19707822

RESUMEN

BACKGROUND: Previously, the authors demonstrated that an intraabdominal pressure (IAP) of 14 mmHg in normal rats reduced kidney function/hemodynamics. These adverse effects are related to interference with the nitric oxide (NO) system. This study was designed to compare the effects of NO synthase (NOS) inhibition on kidney function/hemodynamics during increases in IAP from 0 mmHg to 7, 10, and 14 mmHg. METHODS: The rats were divided into six groups. After an IAP of 0 (baseline), the first three groups were subjected to increasing IAPs as follows: 7 mmHg (group 1), 10 mmHg (group 2), and 14 mmHg (group 3). Each pressure was applied for 1 h, followed by a deflation period of 60 min (recovery). An additional three groups were pretreated with nitro-L: -arginine methyl ester (L: -NAME), an NOS inhibitor, before pressures of 7 mmHg (group 4), 10 mmHg (group 5) and 14 mmHg (group 6) were applied for 1 h. Urine flow rate (V), Na(+) excretion (U(Na)V), glomerular filtration rate (GFR), and renal plasma flow (RPF), were determined throughout the experiments. RESULTS: There were no significant changes in V, U(Na)V, GFR, or RPF during 7-mmHg insufflation. However, significant reductions in these parameters were observed during 10 and 14 mmHg, with V decreasing from 9.95 + or - 1.34 microl/min to 6.8 + or - 1.1 and 6.1 + or - 0.5 microl/min (p < 0.05) and U(Na)V decreasing from 1.29 + or - 0.28 to 0.43 + or - 0.32 muEq/min (p < 0.05), and 0.39 + or - 0.09 muEq/min (p < 0.05). These alterations in excretory functions were associated with considerable declines in GFR, from 1.98 + or - 0.2 to 1.05 + or - 0.18 ml/min (p < 0.05) and 0.95 + or - 0.06 ml/min (p < 0.05) and RPF from 8.66 + or - 0.62 to 3.94 + or - 0.88 ml/min (p < 0.05) and 3.08 + or - 0.71 ml/min (p < 0.05), respectively. When the animals were pretreated with L: -NAME, the adverse renal effects of an IAP of 14 mmHg, but not 10 mmHg, were substantially aggravated. CONCLUSION: Decreased renal function/perfusion is induced by IAP pressures of 10 and 14 mmHg but not 7 mmHg. Inhibition of NOS aggravates the adverse renal effects of high (14 mmHg) but not low (7 or 10 mmHg) IAP, indicating that NO deficiency may contribute to the renal dysfunction during high IAP.


Asunto(s)
Riñón/efectos de los fármacos , Riñón/enzimología , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Neumoperitoneo Artificial/efectos adversos , Análisis de Varianza , Animales , Hemodinámica/efectos de los fármacos , Pruebas de Función Renal , Masculino , Ratas , Ratas Sprague-Dawley
7.
Nephron Physiol ; 115(3): p21-30, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20460939

RESUMEN

BACKGROUND/AIMS: Patients treated with peroxisome proliferator-activated receptor analogs (PPAR) alpha or alpha/gamma may develop a transient and reversible increase in serum creatinine, the mechanism of which remains obscure. This study evaluates whether treatment with either PPAR-alpha or -alpha/gamma analogs, fenofibrate or tesaglitazar, may cause deterioration in renal hemodynamics or exert direct tubular or glomerular nephrotoxic effects in rats. METHODS: Male Sprague-Dawley rats (300-320 g) were treated per os with fenofibrate (300 mg/kg/day), tesaglitazar (1.2 mg/kg/day) or vehicle, for 14 days. Glomerular filtration rate (GFR) and renal blood flow (RBF) were measured by inulin clearance and ultrasonic flowmetry, and cumulative excretion of sodium and creatinine were assessed. Biomarkers of glomerular and tubular injury were measured, including urinary albumin excretion and renal mRNA levels of kidney injury molecule 1 (Kim-1), lipocalin 2 (Lcn2), and osteopontin (Spp1). RESULTS: Fenofibrate and tesaglitazar improved the lipid profile, but caused no detectable decrease in GFR or RBF compared with vehicle-treated rats. Furthermore, the cumulative excretions of sodium and creatinine were not altered by the drugs. Finally, there was no significant difference between drug- and vehicle-treated groups in urinary albumin excretion or in the expression of renal injury biomarkers. CONCLUSIONS: In the rat, no direct nephrotoxic effect or deterioration in renal hemodynamics and function were observed following treatment with fenofibrate or tesaglitazar.


Asunto(s)
Alcanosulfonatos/farmacología , Fenofibrato/farmacología , Túbulos Renales/efectos de los fármacos , PPAR alfa/agonistas , PPAR gamma/agonistas , Fenilpropionatos/farmacología , Alcanosulfonatos/toxicidad , Animales , Moléculas de Adhesión Celular/genética , Creatinina/orina , Fenofibrato/toxicidad , Tasa de Filtración Glomerular/efectos de los fármacos , Tasa de Filtración Glomerular/fisiología , Hipolipemiantes/farmacología , Hipolipemiantes/toxicidad , Inulina/farmacocinética , Túbulos Renales/fisiología , Lipocalina 2 , Lipocalinas/genética , Masculino , Osteopontina/genética , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Fenilpropionatos/toxicidad , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Circulación Renal/efectos de los fármacos , Circulación Renal/fisiología , Sodio/orina
8.
Nephrol Dial Transplant ; 24(2): 428-36, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18799609

RESUMEN

BACKGROUND: Ischaemic kidney injury continues to play a dominant role in the pathogenesis of acute renal failure (ARF) in many surgical and medical settings. A major event in the induction of renal injury is related to the generation of oxygen-free radicals. Hyperbaric oxygen therapy (HBO) is indicated for treatment of many ischaemic events but not for ARF. Therefore, the present study examined the effects of HBO on kidney function and renal haemodynamics in rats with ischaemic ARF. METHODS: Renal ischaemia was induced by unilateral renal artery clamping (45 min) in rats. Within 24 h following ischaemia, rats were treated twice with HBO of 100% O(2) at 2.5 absolute atmospheres for 90 min each (+HBO). Untreated rats (-HBO) served as a control. Forty-eight hours later, GFR, RBF and endothelial-dependent vasorelaxation were measured. In addition, the immunoreactive staining of 4-hydroxy-2-noneal (4-HNE), a major product of endogenous lipid peroxidation, and superoxide dismutase (SOD) were assessed. RESULTS: In the -HBO group, GFR was reduced by 94% compared with the untouched normal kidney (ischaemic: 0.06 +/- 0.03 ml/min, normal: 1.02 +/- 0.13 ml). In contrast, in the +HBO group, GFR of the ischaemic kidney (0.36 +/- 0.07 ml/min) was reduced only by 68% compared with the contralateral normal kidney (1.12 +/- 0.12 ml/min). In line with these findings, HBO improved the vasodilatory response to ACh as expressed in enhancement of both total and regional renal blood flow. In addition, HBO reduced the formation of 4-HNE by 33% and 76% and increased SOD by 30% and 70% in the cortex and outer stripe region of the medulla of the ischaemic kidney, respectively. CONCLUSION: HBO attenuates the decline in GFR following renal ischaemia, and improves endothelial-dependent vasorelaxation, suggesting that treatment with HBO may be beneficial in the setting of ischaemic ARF.


Asunto(s)
Tasa de Filtración Glomerular , Oxigenoterapia Hiperbárica , Riñón/irrigación sanguínea , Riñón/lesiones , Daño por Reperfusión/terapia , Lesión Renal Aguda/fisiopatología , Lesión Renal Aguda/terapia , Animales , Antioxidantes/metabolismo , Endotelio Vascular/fisiopatología , Isquemia/metabolismo , Isquemia/fisiopatología , Riñón/fisiopatología , Peroxidación de Lípido , Masculino , Oxidantes/metabolismo , Ratas , Ratas Sprague-Dawley , Circulación Renal , Daño por Reperfusión/fisiopatología , Superóxido Dismutasa/metabolismo , Vasodilatación
9.
Kidney Blood Press Res ; 32(4): 293-303, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19797914

RESUMEN

BACKGROUND/AIMS: Endothelin (ET)-1 is produced by most renal cell types. Renal tubular and vascular cells express both the ET receptors ET(A) and ET(B). Since significant amounts of ET-1 of renal origin were detected in human urine, urinary ET-1 has been used as an index for the capacity of renal ET-1 production. Here, we determine the existence of additional components of the intrarenal ET system, namely the ET(A) and ET(B) receptor subtypes, in the urine of normal and hypertensive subjects. METHODS: ET(A) and ET(B) receptors were detected in urine samples that were concentrated by TCA precipitation, Speedvac or ProteoSpin. RESULTS: Analysis of the human urine extracts revealed the existence of approximately 50 and 55 kDa of immunoreactive proteins, corresponding to ET(B) and ET(A), respectively, indicating that intact ET(A) and ET(B) are excreted in the urine of healthy subjects and hypertensive patients. Normotensive and hypertensive subjects had statistically comparable ET(B) excretion normalized to creatinine (0.58 +/- 0.16 vs. 0.83 +/- 0.17 microg/mg creatinine, respectively; p = 0.304). In contrast, ET(A) excretion was higher among hypertensive subjects (0.05 +/- 0.01 vs. 0.11 +/- 0.02 microg/mg creatinine; p = 0.0451). Immunostaining of ET(A) and ET(B) in the human urinary system revealed expression of both receptors, principally in tubular cells (mainly in medullary collecting ducts) and in the bladder urothelium, and ET(A) expression in the peritubular capillaries and arterioles. Urinary ET receptors closely and inversely correlated with indices of urine concentration, suggesting that their shedding is principally affected by urine flow. CONCLUSION: ET receptors are present in human urine, conceivably originating within the urinary system. Their excretion is principally affected by urinary concentration. It remains to be determined whether urinary ET(A)/ET(B) is of physiological/pathophysiological relevance.


Asunto(s)
Hipertensión/metabolismo , Riñón/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Western Blotting , Creatinina/orina , Femenino , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Humanos , Hipertensión/patología , Inmunoprecipitación , Riñón/patología , Corteza Renal/patología , Médula Renal/patología , Masculino , Persona de Mediana Edad , Análisis de Regresión , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Ácido Tricloroacético/química
10.
J Pharmacol Exp Ther ; 326(2): 414-22, 2008 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-18467593

RESUMEN

Arginine vasopressin (AVP) plays an important role in renal hemodynamic alterations, water retention, and cardiac remodeling in congestive heart failure (CHF). The present study evaluated the acute and chronic effects of vasopressin V(1a) receptor subtype (V(1a)) and vasopressin V(2) receptor subtype (V(2)) antagonists on renal function and cardiac hypertrophy in rats with CHF. The effects of acute administration of SR 49059 [(2S)1-[(2R,3S)-5-chloro-3-(2-chlorophenyl)-1-(3,4-dimethoxybenzene-sulfonyl)-3-hydroxy-2,3-dihydro-1H-indole-2-carbonyl]-pyrrolidine-2-carboxamide)] (0.1 mg/kg) and SR 121463B (1-[4-(N-tert-butylcarbamoyl)-2-methoxybenzenesulfonyl]-5-ethoxy-3-spiro-[4-(2-morpholinoethoxy)cyclohexane]indol-2-one, fumarate; equatorial isomer) (0.3 mg/kg), V(1a) and V(2) antagonists, respectively, on renal function, and of chronic treatment (3.0 mg/kg/day for 7 or 28 days, via osmotic minipumps or p.o.), on water excretion and cardiac hypertrophy were studied in rats with aortocaval fistula and control rats. CHF induction increased plasma AVP (12.8 +/- 2.5 versus 32.2 +/- 8.3 pg/ml, p < 0.05). Intravenous bolus injection of SR 121463B to controls produced dramatic diuretic response (from 5.5 +/- 0.8 to 86.3 +/- 21.9 microl/min; p < 0.01). In contrast, administration of SR 49059 did not affect urine flow. Likewise, administration of SR 121463B, but not SR 49059, to rats with CHF significantly increased urinary flow rate from 20.8 +/- 6.4 to 91.6 +/- 26.5 microl/min (p < 0.01). The diuretic effects of SR 121463B were associated with a significant decline in urinary osmolality and insignificant change of Na+ excretion. In line with its acute effects, chronic administration of SR 121463B to CHF rats increased daily urinary volume 2 to 5-fold throughout the treatment period. Both SR 121463B and SR 49059 significantly reduced heart weight in CHF rats when administered for 4 weeks, but not 1 week. These results suggest that V(2) and V(1a) antagonists improve water balance and cardiac hypertrophy in CHF and might be beneficial for the treatment of water retention and cardiac remodeling in CHF.


Asunto(s)
Antagonistas de los Receptores de Hormonas Antidiuréticas , Cardiomegalia/tratamiento farmacológico , Diuresis/efectos de los fármacos , Insuficiencia Cardíaca/tratamiento farmacológico , Indoles/uso terapéutico , Morfolinas/uso terapéutico , Pirrolidinas/uso terapéutico , Compuestos de Espiro/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Cardiomegalia/complicaciones , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Esquema de Medicación , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/fisiopatología , Indoles/administración & dosificación , Indoles/farmacología , Pruebas de Función Renal , Masculino , Morfolinas/administración & dosificación , Morfolinas/farmacología , Tamaño de los Órganos/efectos de los fármacos , Concentración Osmolar , Pirrolidinas/administración & dosificación , Pirrolidinas/farmacología , Ratas , Ratas Sprague-Dawley , Sodio/orina , Compuestos de Espiro/administración & dosificación , Compuestos de Espiro/farmacología , Vasoconstricción/efectos de los fármacos
11.
EXS ; (95): 123-32, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16383002

RESUMEN

The presence in the mammalian kidney of NPY and at least one of its receptor subtypes has been proven by several independent methodologies. Also, numerous studies using physiological and pharmacological approaches indicated that this peptide has the capacity to alter renal function. In particular, these studies suggest that NPY may exert renal vasoconstrictor and tubular actions that are species dependent, and may also influence renin secretion by the kidney. The question whether NPY plays an important role in the physiological regulation of renal hemodynamics and electrolyte excretion, remains largely unanswered at present. No major impairments in renal function have been reported in genetically models deficient in NPY or its Y1 receptor. Thus, additional studies are required to elucidate the role of NPY in the physiological and pathophysiological regulation of renal function.


Asunto(s)
Enfermedades Renales/metabolismo , Enfermedades Renales/fisiopatología , Riñón/fisiología , Neuropéptido Y/fisiología , Animales , Humanos , Riñón/fisiopatología , Pruebas de Función Renal , Neuropéptido Y/genética , Receptores de Neuropéptido Y/fisiología
12.
Pharmacol Ther ; 168: 75-97, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27598860

RESUMEN

The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.


Asunto(s)
Insuficiencia Cardíaca/etiología , Hipoglucemiantes/efectos adversos , Tiazolidinedionas/efectos adversos , Animales , Líquidos Corporales/efectos de los fármacos , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Edema/inducido químicamente , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipoglucemiantes/administración & dosificación , PPAR gamma/agonistas , Factores de Riesgo , Tiazolidinedionas/administración & dosificación
13.
Circulation ; 105(2): 236-43, 2002 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-11790707

RESUMEN

BACKGROUND: Whereas nitric oxide (NO) has been implicated in the pathophysiology of heart failure (HF), the significance and functional role of different NO synthase (NOS) isoforms in this pathology are controversial. Our aim was to study in the myocardium of rats with volume-overload-induced HF the expression, activity, and localization of endothelial (eNOS) and inducible (iNOS) isoforms and the involvement of iNOS in depressed cardiac contractile properties, intracellular Ca(2+) ([Ca(2+)](i)) transients, and beta-adrenergic hyporesponsiveness. METHODS AND RESULTS: HF was induced by aortocaval fistula (ACF). Compensated and decompensated subgroups of HF were selected on the basis of daily sodium excretion. ACF induced cardiac hypertrophy in rats with compensated (36%) and decompensated (76%) HF. Whereas in HF rats, cardiac eNOS expression and activity were unchanged, iNOS expression and activity increased approximately 2-fold. iNOS immunostaining was observed in ventricular myocytes of compensated and decompensated HF rats but not in controls. Isoproterenol-positive inotropic and lusitropic effects were markedly attenuated in papillary muscle of HF rats, more pronouncedly in decompensated than in compensated rats. Isoproterenol-induced increases in the rates of [Ca(2+)](i) activation and relaxation were also depressed in ACF rats. Selective iNOS blockade by N-(3-(aminomethyl)benzylacetamidine improved the attenuated beta-adrenergic responsiveness in HF rats. CONCLUSIONS: Our findings indicate that myocardial iNOS is activated in rats with volume-overload HF and suggest that increased iNOS activity contributes to depressed myocardial contractility and beta-adrenergic hyporesponsiveness.


Asunto(s)
Agonistas Adrenérgicos beta/farmacología , Insuficiencia Cardíaca/enzimología , Insuficiencia Cardíaca/fisiopatología , Isoproterenol/farmacología , Contracción Miocárdica , Óxido Nítrico Sintasa/fisiología , Amidinas/farmacología , Animales , Fístula Arteriovenosa/complicaciones , Bencilaminas/farmacología , Calcio/metabolismo , Volumen Cardíaco , Cardiomegalia/patología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Insuficiencia Cardíaca/etiología , Ventrículos Cardíacos/citología , Ventrículos Cardíacos/metabolismo , Contracción Miocárdica/efectos de los fármacos , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Óxido Nítrico Sintasa de Tipo III , Músculos Papilares/efectos de los fármacos , Músculos Papilares/enzimología , Ratas
14.
Pharmacol Ther ; 102(3): 223-41, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15246247

RESUMEN

The natriuretic peptide family consists of at least 3 structurally similar peptides: atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), and C-type natriuretic peptide (CNP). Under normal conditions, ANP is synthesized by the atrium and released in response to atrial stretch. This peptide plays an important role in sodium and water homeostasis and is involved in cardiovascular function. In contrast, BNP is synthesized primarily by the ventricles, and its circulatory concentrations are significantly elevated in profound congestive heart failure (CHF). While both plasma levels of ANP and BNP have been found to be increased in patients with various heart diseases, the elevation in circulatory BNP correlates better than ANP with the severity of CHF. Therefore, plasma BNP has been suggested (and lately used) to aid in the accurate diagnosis of heart failure in patients admitted to the emergency room with symptoms of decompensated heart failure. Furthermore, circulatory BNP has been utilized as a prognostic marker in CHF as well as a hormone guide in the evaluation of the efficacy of the conventional treatment of this disease state. In light of the cardiovascular and renal effects of BNP, which most likely exceed those of ANP, the former has been used as a therapeutic agent for the treatment of patients with acute severe CHF. Intravenous infusion of BNP into patients with sustained ventricular dysfunction causes a balanced arterial and venous vasodilatation that has been shown to result in rapid reduction in ventricular filling pressure and reversal of heart failure symptoms, such as dyspnea and acute hemodynamic abnormalities. Thus, the goal of this article is to review the physiology and pathophysiology of natriuretic peptides and the potential use of their circulating levels for diagnosis and treatment of heart failure.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Péptidos Natriuréticos/fisiología , Aldosterona/fisiología , Animales , Factor Natriurético Atrial/fisiología , Factor Natriurético Atrial/uso terapéutico , Endopeptidasas/fisiología , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/fisiopatología , Humanos , Péptido Natriurético Encefálico/fisiología , Péptido Natriurético Encefálico/uso terapéutico , Péptido Natriurético Tipo-C/fisiología , Péptido Natriurético Tipo-C/uso terapéutico , Péptidos Natriuréticos/uso terapéutico , Inhibidores de Proteasas/uso terapéutico , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/fisiología
15.
J Invest Surg ; 18(6): 325-34, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16319054

RESUMEN

Much of the understanding about the pathophysiological responses to chronic cardiac overload has been gained by the use of rat and dog models of aortocaval fistula (ACF). The use of a similar model in genetically manipulated mice may further elucidate the molecular mechanisms in these responses. The only reports about ACF in mice to date have applied a needle puncture to create the ACF, which may result in an uncontrolled and irreproducible size of the shunt, and require several weeks to induce the characteristic cardiac changes. In order to obtain a more consistent approach to characterize this mode of cardiac hyperfunction, we present a surgical murine model of ACF that results in rapid progression of the typical systemic and cardiac changes. A sutureless side-to-side infrarenal surgical anastomosis of 0.6-0.8 mm in diameter was created between the abdominal aorta and inferior vena cava in ICR (Institute of Cancer Research) mice. Six to 7 days later, significant cardiac hypertrophy developed. The heart/body weight ratio increased from 0.45 +/- 0.02% in control mice to 0.77 +/- 0.03% in mice with ACF (p < .003). The dry heart weight ratio increased from 0.099 +/- 0.0033% to 0.13 +/- 0.008% (p < .006). The ACF dramatically induced the atrial and ventricular expression of atrial natriuretic factor mRNA, and increased the total cardiac content of endothelin-1 (162.5 +/- 50.6 vs. 83.9 +/- 9.0 pg). Mean arterial pressure in anesthetized mice with ACF decreased from 69.8 +/- 4.9 to 54.8 +/- 5.5 mm Hg (p < .025). Urinary sodium excretion returned to preoperative levels several days following surgery. These results demonstrate that cardiac hypertrophy could be rapidly and reproducibly achieved in mice by the placement of a surgical ACF. This model, when applied in genetically manipulated mice, may be a valuable tool for functional genomic studies about the pathogenesis of cardiac hypertrophy and heart failure.


Asunto(s)
Aorta Abdominal/cirugía , Derivación Arteriovenosa Quirúrgica , Cardiomegalia/etiología , Modelos Animales de Enfermedad , Vena Cava Inferior/cirugía , Animales , Factor Natriurético Atrial/biosíntesis , Presión Sanguínea , Northern Blotting , Endotelina-1/biosíntesis , Ratones , Ratones Endogámicos ICR , Miocardio/metabolismo
16.
Curr Vasc Pharmacol ; 1(3): 347-54, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15320481

RESUMEN

Large Arteriovenous Fistulae (AVF) can increase cardiac output. This may result in the development of congestive heart failure, a clinical situation associated with increased activity of Vasoconstrictor neurohormonal systems: the Renin-angiotensin system (RAS), Sympathetic nervous system (SNS), the Endothelin system and Arginine vasopressin (AVP). At the same time there is compensatory activation of systemic and vasodilating systems: Atrial natriuretic peptide (ANP) and Nitric oxide (NO). Previous data from our laboratory and other groups suggest that urinary sodium excretion in this situation is largely determined by the balance between two antagonistic hormonal systems: the vasoconstrictor/sodium-retaining factors such as RAS, endothelin, and SNS, and vasodilatory/natriuretic substances such as ANP and NO. In decompensated patients, enhanced activities of the sodium-retaining systems overwhelm the effects of vasodilatory/natriuretic systems, which lead to a net reduction in sodium and water excretion. For compensation to occur, the effects of natriuretic mechanisms must prevail over those of the opposing systems, resulting in renal sodium/water excretion. This notion is supported by clinical and experimental studies where pharmacological intervention corrected the imbalance present in AVF. Thus, a shift in the balance in favor of natriuresis may be achieved either by increasing the activity of natriuretic factors or reducing the influence of the antinatriuretic systems. Based on that, the use of angiotensin converting enzyme (ACE) inhibitors and/or angiotensin II (ATII) blockers may be beneficial in the management of patients with large AVF.


Asunto(s)
Fístula Arteriovenosa/fisiopatología , Angiotensina II/antagonistas & inhibidores , Bloqueadores del Receptor Tipo 1 de Angiotensina II/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , Animales , Arginina Vasopresina/fisiología , Fístula Arteriovenosa/complicaciones , Fístula Arteriovenosa/tratamiento farmacológico , Factor Natriurético Atrial/fisiología , Endotelinas/fisiología , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/metabolismo , Humanos , Óxido Nítrico/fisiología , Sistema Renina-Angiotensina/fisiología , Sistema Nervioso Simpático/fisiología
17.
J Cardiovasc Pharmacol ; 44 Suppl 1: S362-5, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15838321

RESUMEN

Endothelin-1 (ET-1) exerts its biological actions through two receptor subtypes: endothelin-A (ETA) receptor and endothelin-B (ETB) receptor. We demonstrated previously that ET-1 induces systemic and renal cortical vasoconstriction via ETA whereas ETB mediates medullary vasodilation. Congestive heart failure (CHF) is characterized by increased vascular resistance and impaired renal hemodynamic and excretory function. While the pathophysiological effects of ET-1 in CHF are well established, the status of ETA and ETB in the kidney is poorly characterized. The present study examined the immunostaining and localization of ETA and ETB in the renal cortex and medulla of rats with experimental CHF induced by aorto-caval fistula. Rats with CHF were further subdivided, based on their daily urinary sodium excretion, into rats with compensated (urinary sodium excretion > 1200 microEq/day) and decompensated CHF (urinary sodium excretion < 200 microEq/day). ETA is predominantly localized to the cortex mainly in the peritubular capillaries, and is upregulated in rats with compensated and decompensated CHF compared with sham controls. In contrast, ETB is preferentially expressed in the outer and inner medulla, mainly in the vasa recta, the thick ascending limb of Henle's loop and the collecting duct. While compensated CHF is associated with upregulation of ETB in the collecting duct and vasa recta, decompensated CHF is accompanied with enhanced ETB abundance in the vasa recta and remarkable downregulation of this receptor subtype in the collecting duct. The findings suggest that upregulation of ETA may lead to a decrease in cortical blood flow while upregulation of ETB in the vasa recta probably contributes to the preservation of medullary blood flow. Furthermore, downregulation of ETB in the collecting duct, only in rats with decompensated CHF, could contribute to sodium retention in that subgroup.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Corteza Renal/metabolismo , Médula Renal/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Animales , Aorta Abdominal/cirugía , Derivación Arteriovenosa Quirúrgica , Modelos Animales de Enfermedad , Insuficiencia Cardíaca/fisiopatología , Inmunohistoquímica , Corteza Renal/irrigación sanguínea , Médula Renal/irrigación sanguínea , Masculino , Natriuresis , Ratas , Ratas Wistar , Circulación Renal , Vena Cava Inferior/cirugía
18.
J Cardiovasc Pharmacol ; 44 Suppl 1: S54-8, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15838359

RESUMEN

Endothelin-1 (ET-1) is involved in the pathogenesis of cardiac and renal hemodynamic changes and impaired excretory function in congestive heart failure. It has previously been demonstrated that acute administration of ABT-627 (endothelin-A blocker) abolished systemic and renal vasoconstriction in controls and rats with congestive heart failure induced by a surgically created aortocaval fistula (Abassi et al. Clin Sci (Lond) 2002;103:S245-S248). In contrast, acute endothelin-B blockade by A-192621 exaggerated the ET-1 induced systemic and renal vasoconstriction. The present study examined the renal and systemic effects of chronically administered ABT-627 (24 mg/kg per day) or A-192621 (72 mg/kg per day) for 7 days via osmotic minipumps inserted intraperitoneally on the day of operation of sham controls and rats with congestive heart failure. Tailcuff measurements revealed that ABT- 627 significantly decreased mean arterial pressure from 108 +/- 2 mmHg to 87 +/- 2 mmHg (P < 0.05), whereas A-192621 significantly increased mean arterial pressure from 110 +/- 3 mmHg to 122 +/- 3 mmHg (P < 0.05) in controls. Despite the hypotensive effect of ABT-627, daily sodium excretion dramatically increased, but to a lesser extent in A-192621-treated controls. Furthermore, chronic administration of ABT-627 to controls attenuated the systemic and renal vasoconstriction induced by ET-1 (1 nmol/kg intravenous), whereas A-192621 augmented these effects. Similarly, chronic treatment with ABT-627 totally abolished the systemic and renal vasoconstriction caused by injected ET-1 in rats with congestive heart failure, whereas A192621 potentiated these effects. Chronic treatment of animals with congestive heart failure with ABT-627 did not influence daily sodium excretion, whereas treatment with A192621 significantly improved daily sodium excretion. Interestingly, treatment with either ABT-627 or A192621 significantly decreased cardiac hypertrophy in rats with congestive heart failure. In conclusion, in sham controls endothelin-B receptor mediated vasodilation and natriuresis, probably as a result of tubular action, whereas in congestive heart failure the excretory contribution of endothelin-B receptor was attenuated, resulting in Na+ retention.


Asunto(s)
Antagonistas de los Receptores de la Endotelina A , Antagonistas de los Receptores de la Endotelina B , Insuficiencia Cardíaca/tratamiento farmacológico , Hemodinámica/efectos de los fármacos , Riñón/efectos de los fármacos , Pirrolidinas/farmacología , Animales , Atrasentán , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/etiología , Cardiomegalia/prevención & control , Modelos Animales de Enfermedad , Endotelina-1/metabolismo , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/fisiopatología , Bombas de Infusión Implantables , Riñón/metabolismo , Riñón/fisiopatología , Masculino , Natriuresis/efectos de los fármacos , Pirrolidinas/administración & dosificación , Ratas , Ratas Wistar , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Factores de Tiempo , Vasoconstricción/efectos de los fármacos , Vasodilatación/efectos de los fármacos
19.
J Cardiovasc Pharmacol ; 44 Suppl 1: S191-4, 2004 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-15838276

RESUMEN

The Cohen-Rosenthal diabetic hypertensive rat is a unique animal model in which genetic hypertension and diabetes developed after crossbreeding of a sensitive substrain of Cohen diabetic rats and spontaneously hypertensive rats (SHR) and feeding them a copper-poor sucrose diet. This study examined the acute effects of endothelin-1 on the systemic and renal hemodynamics in Cohen-Rosenthal diabetic hypertensive rats, Cohen diabetic rats and spontaneously hypertensive rats. Intravenous injection of endothelin- 1 (1.0 nmol/kg) into anesthetized SHR resulted in a significant immediate depressor response in mean arterial pressure [from 165 +/- 3 mmHg to 124 +/- 12 mmHg (P < 0.0001)] followed by a minor hypertensive phase (mean arterial pressure increased to 170 +/- 2 mmHg). Simultaneously, the administration of endothelin-1 caused a significant decrease in renal blood flow from 5.8 +/- 0.9 mL/minute to 3.2 +/- 0.5 mL/minute (P = 0.026). These responses were blunted in Cohen-Rosenthal diabetic hypertensive rats and Cohen diabetic rats. Analysis of intrarenal blood flow by laser-Doppler in Cohen-Rosenthal diabetic hypertensive rats revealed that endothelin-1 injection caused a decrease in cortical blood flow (Delta = -12 +/- 2.9%). However, in contrast to its well known renal medullary vasodilatory effect, endothelin-1 produced a significant decline in the medulla blood flow (Delta = -17.5 +/- 3.4%) (P = 0.0125). These findings suggest that Cohen diabetic rats and Cohen-Rosenthal diabetic hypertensive rats have reduced sensitivity to the vascular and renal action of endothelin-1. Furthermore, in the Cohen-Rosenthal diabetic hypertensive rats the expected endothelin- 1-induced medullary vasodilation was abolished and even reversed into prolonged vasoconstrictor response.


Asunto(s)
Diabetes Mellitus/metabolismo , Endotelina-1/metabolismo , Hemodinámica , Riñón/irrigación sanguínea , Circulación Renal , Animales , Presión Sanguínea , Cobre/deficiencia , Diabetes Mellitus/etiología , Diabetes Mellitus/fisiopatología , Sacarosa en la Dieta , Modelos Animales de Enfermedad , Endotelina-1/administración & dosificación , Inyecciones Intravenosas , Ratas , Ratas Endogámicas SHR , Resistencia Vascular , Vasoconstricción , Vasodilatación
20.
Physiol Rep ; 1(6): e00155, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24400157

RESUMEN

Xanthurenic acid 8-o-ß-d-glucoside is an endogenous derivative of tryptophan metabolism, isolated from urine of patients with chronic renal disease. This compound was suggested previously to act as a natriuretic hormone based on its ability to block short circuit currents in a frog skin assay and to induce a sustained natriuresis when injected into rats (C. D. Cain et al., Proc. Natl. Acad. Sci. USA 2007: 17873-17878). The present communication describes the effects of the compound on renal clearance and hemodynamic parameters in male Sprague-Dawley rats maintained on a normal salt (0.4-0.5%) diet. Intravenous administration of synthetic xanthurenic acid 8-o-ß-d-glucoside in two consecutive incremental doses (6.3 and 31.5 nmol) resulted in a significant increase (P < 0.05), in urine flow (43.91 ± 6.31 µL/min vs. 10.54 ± 2.21 µL/min), absolute rate of sodium excretion (3.99 ± 0.95 µEq/min vs. 1.15 ± µEq/min), and percentage sodium excretion (1.63 ± 0.46% vs. 0.37 ± 0.12%, peak response vs. baseline, respectively). The natriuretic/diuretic effect was associated also with a significant increase in potassium excretion. These effects were not related to changes in renal hemodynamics or in arterial blood pressure. Pretreatment with the sodium channel blocker, amiloride, completely abolished the natriuretic and kaluretic actions of the compound. Administration of the xanthurenic acid derivative caused a dose-related increase in urinary nitrite/nitrate excretion. Moreover, under chronic nitric oxide blockade by l-NG-Nitro-Arginine-Methyl-Esther (l-NAME) sodium excretion was similar in rats treated or untreated with the compound. Our data demonstrate that xanthurenic acid 8-o-ß-d-glucoside has significant diuretic/natriuretic and kaluretic properties. An intact amiloride-sensitive sodium channel is required for the renal effects of the compound. The data further suggest that the natriuretic effect is mediated in part by a nitric oxide-dependent mechanism.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA