Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Int J Mol Sci ; 24(6)2023 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-36982455

RESUMEN

Soluble fms-like tyrosine kinase-1 (sFlt-1) is a secreted protein that binds heparan sulfate expressed on the endothelial glycocalyx (eGC). In this paper we analyze how excess sFlt-1 causes conformational changes in the eGC, leading to monocyte adhesion, a key event triggering vascular dysfunction. In vitro exposure of primary human umbilical vein endothelial cells to excess sFlt-1 decreased eGC height and increased stiffness as determined by atomic force microscopy (AFM). Yet, structural loss of the eGC components was not observed, as indicated by Ulex europaeus agglutinin I and wheat germ agglutinin staining. Moreover, the conformation observed under excess sFlt-1, a collapsed eGC, is flat and stiff with unchanged coverage and sustained content. Functionally, this conformation increased the endothelial adhesiveness to THP-1 monocytes by about 35%. Heparin blocked all these effects, but the vascular endothelial growth factor did not. In vivo administration of sFlt-1 in mice also resulted in the collapse of the eGC in isolated aorta analyzed ex vivo by AFM. Our findings show that excess sFlt-1 causes the collapse of the eGC and favors leukocyte adhesion. This study provides an additional mechanism of action by which sFlt-1 may cause endothelial dysfunction and injury.


Asunto(s)
Preeclampsia , Receptor 1 de Factores de Crecimiento Endotelial Vascular , Humanos , Animales , Ratones , Femenino , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Glicocálix/metabolismo , Endotelio/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Preeclampsia/metabolismo
2.
J Am Soc Nephrol ; 32(8): 1853-1863, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34155060

RESUMEN

Soluble Fms-like tyrosine kinase (sFlt-1/sVEGFR1) is a naturally occurring antagonist of vascular endothelial growth factor (VEGF). Despite being a secreted, soluble protein lacking cytoplasmic and transmembrane domains, sFlt-1 can act locally and be protective against excessive microenvironmental VEGF concentration or exert autocrine functions independently of VEGF. Circulating sFlt-1 may indiscriminately affect endothelial function and the microvasculature of distant target organs. The clinical significance of excess sFlt-1 in kidney disease was first shown in preeclampsia, a major renal complication of pregnancy. However, circulating sFlt-1 levels appear to be increased in various diseases with varying degrees of renal impairment. Relevant clinical associations between circulating sFlt-1 and severe outcomes (e.g., endothelial dysfunction, renal impairment, cardiovascular disease, and all-cause mortality) have been observed in patients with CKD and after kidney transplantation. However, sFlt-1 appears to be protective against renal dysfunction-associated aggravation of atherosclerosis and diabetic nephropathy. Therefore, in this study, we provide an update on sFlt-1 in several kidney diseases other than preeclampsia, discuss clinical findings and experimental studies, and briefly consider its use in clinical practice.


Asunto(s)
Lesión Renal Aguda/sangre , Microvasos/patología , Insuficiencia Renal Crónica/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Lesión Renal Aguda/cirugía , Biomarcadores/sangre , Vasos Coronarios/patología , Endotelio/fisiopatología , Femenino , Humanos , Riñón/irrigación sanguínea , Riñón/patología , Trasplante de Riñón , Preeclampsia/sangre , Embarazo , Diálisis Renal , Insuficiencia Renal Crónica/terapia
3.
Kidney Int ; 95(5): 1091-1102, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30824181

RESUMEN

Renal ischemia reperfusion injury (IRI) adversely affects clinical outcomes following kidney transplantation. Understanding the cellular mechanisms and the changes in gene/protein expression following IRI may help to improve these outcomes. Serum soluble fms-like tyrosine kinase 1 (sFlt-1), a circulating antiangiogenic protein, is increased in the first week following kidney transplantation. We evaluated the casual relationship of elevated sFlt-1 levels with renal microvascular dysfunction following IRI in a longitudinal study of 93 kidney transplant recipients and in several animal models. Transplant recipients with higher sFlt-1 levels had higher odds of delayed graft function, graft rejection, impaired graft function, and death. In a subgroup of 25 participants who underwent kidney biopsy within 4 months of kidney transplantation, peritubular capillary area was lower in those with elevated serum sFtl-1 levels. The administration of recombinant sFlt-1 into rodents resulted in significant structural and functional changes of the renal microvasculature, including reduced peritubular capillary density and intracapillary blood volume, and lead to increased expression of inflammatory genes and increased fibrosis. In a murine model of IRI, the kidney was a site of sFlt-1 production, and systemic neutralization of sFlt-1 preserved peritubular capillary density and alleviated renal fibrosis. Our data indicate that high sFlt-1 levels after IRI play an important role in the pathogenesis of microvascular dysfunction, thereby contributing to adverse clinical outcomes following kidney transplantation.


Asunto(s)
Fallo Renal Crónico/cirugía , Trasplante de Riñón/efectos adversos , Daño por Reperfusión/sangre , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Adulto , Anciano , Aloinjertos/irrigación sanguínea , Aloinjertos/patología , Animales , Biopsia , Capilares/patología , Línea Celular , Estudios de Cohortes , Funcionamiento Retardado del Injerto/sangre , Funcionamiento Retardado del Injerto/etiología , Funcionamiento Retardado del Injerto/mortalidad , Modelos Animales de Enfermedad , Femenino , Fibrosis , Rechazo de Injerto/sangre , Rechazo de Injerto/etiología , Rechazo de Injerto/mortalidad , Humanos , Riñón/irrigación sanguínea , Riñón/patología , Fallo Renal Crónico/mortalidad , Estudios Longitudinales , Masculino , Ratones , Persona de Mediana Edad , Proteínas Recombinantes/administración & dosificación , Daño por Reperfusión/etiología , Daño por Reperfusión/mortalidad , Resultado del Tratamiento , Receptor 1 de Factores de Crecimiento Endotelial Vascular/administración & dosificación
4.
Kidney Int ; 90(5): 985-996, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27457912

RESUMEN

Patients with chronic kidney disease (CKD) develop increased levels of the phosphate-regulating hormone, fibroblast growth factor (FGF) 23, that are associated with a higher risk of mortality. Increases in inflammatory markers are another common feature that predicts poor clinical outcomes. Elevated FGF23 is associated with higher circulating levels of inflammatory cytokines in CKD, which can stimulate osteocyte production of FGF23. Here, we studied whether FGF23 can directly stimulate hepatic production of inflammatory cytokines in the absence of α-klotho, an FGF23 coreceptor in the kidney that is not expressed by hepatocytes. By activating FGF receptor isoform 4 (FGFR4), FGF23 stimulated calcineurin signaling in cultured hepatocytes, which increased the expression and secretion of inflammatory cytokines, including C-reactive protein. Elevating serum FGF23 levels increased hepatic and circulating levels of C-reactive protein in wild-type mice, but not in FGFR4 knockout mice. Administration of an isoform-specific FGFR4 blocking antibody reduced hepatic and circulating levels of C-reactive protein in the 5/6 nephrectomy rat model of CKD. Thus, FGF23 can directly stimulate hepatic secretion of inflammatory cytokines. Our findings indicate a novel mechanism of chronic inflammation in patients with CKD and suggest that FGFR4 blockade might have therapeutic anti-inflammatory effects in CKD.


Asunto(s)
Citocinas/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Hepatocitos/metabolismo , Inflamación/metabolismo , Insuficiencia Renal Crónica/metabolismo , Animales , Calcineurina/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/metabolismo , Humanos , Proteínas Klotho , Ratones , Factores de Transcripción NFATC/metabolismo , Fosfolipasa C gamma/metabolismo , Cultivo Primario de Células , Ratas , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
5.
Basic Res Cardiol ; 110(3): 30, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25893874

RESUMEN

Chronic kidney disease (CKD) is associated with an increased risk of heart failure (HF). Elevated plasma concentrations of soluble Flt-1 (sFlt-1) have been linked to cardiovascular disease in CKD patients, but whether sFlt-1 contributes to HF in CKD is still unknown. To provide evidence that concludes a pathophysiological role of sFlt-1 in CKD-associated HF, we measured plasma sFlt-1 concentrations in 586 patients with angiographically documented coronary artery disease and renal function classified according to estimated glomerular filtration rate (eGFR). sFlt-1 concentrations correlated negatively with eGFR and were associated with signs of heart failure, based on New York Heart Association functional class and reduced left ventricular ejection fraction (LVEF), and early mortality. Additionally, rats treated with recombinant sFlt-1 showed a 15 % reduction in LVEF and a 29 % reduction in cardiac output compared with control rats. High sFlt-1 concentrations were associated with a 15 % reduction in heart capillary density (number of vessels/cardiomyocyte) and a 24 % reduction in myocardial blood volume. Electron microscopy and histological analysis revealed mitochondrial damage and interstitial fibrosis in the hearts of sFlt-1-treated, but not control rats. In 5/6-nephrectomised rats, an animal model of CKD, sFlt-1 antagonism with recombinant VEGF121 preserved heart microvasculature and significantly improved heart function. Overall, these findings suggest that a component of cardiovascular risk in CKD patients could be directly attributed to sFlt-1. Assessment of patients with CKD confirmed that sFlt-1 concentrations were inversely correlated with renal function, while studies in rats suggested that sFlt-1 may link microvascular disease with HF in CKD.


Asunto(s)
Insuficiencia Cardíaca/etiología , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/metabolismo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Masculino , Microvasos/patología , Ratas , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Eur Heart J ; 32(15): 1935-45, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21138940

RESUMEN

AIMS: Chronic kidney disease is directly associated with cardiovascular complications. Heart remodelling, including fibrosis, hypertrophy, and decreased vascularization, is frequently present in renal diseases. Our objective was to investigate the impact of calcineurin inhibitors (CNI) on cardiac remodelling and function in a rat model of renal disease. METHODS AND RESULTS: Male Sprague Dawley rats were divided into six groups: sham-operated rats, 5/6 nephrectomized rats (Nx) treated with vehicle, CNI (cyclosporine A 5.0 or 7.5, or tacrolimus 0.5 mg/kg/day) or hydralazine (20 mg/kg twice a day) for 14 days, starting on the day of surgery. Creatinine clearance was significantly lower and blood pressure significantly higher in Nx rats when compared with controls. Morphological and echocardiographic analyses revealed increased left ventricular hypertrophy and decreased number of capillaries in Nx rats. Treatment with CNI affected neither the renal function nor the blood pressure, but prevented the development of cardiac hypertrophy and improved vascularization. In addition, regional blood volume improved as confirmed by contrast agent-based echocardiography. Hydralazine treatment did not avoid heart remodelling in this model. Gene expression analysis verified a decrease in hypertrophic genes in the heart of CNI-treated rats, while pro-angiogenic and stem cell-related genes were upregulated. Moreover, mobilization of stem/progenitor cells was increased through manipulation of the CD26/SDF-1 system. CONCLUSION: We conclude from our studies that CNI-treatment significantly prevented cardiac remodelling and improved heart function in Nx rats without affecting renal function and blood pressure. This sheds new light on possible therapeutic strategies for renal patients at high cardiovascular risk.


Asunto(s)
Inhibidores de la Calcineurina , Ciclosporina/uso terapéutico , Cardiopatías/prevención & control , Inmunosupresores/uso terapéutico , Enfermedades Renales/complicaciones , Tacrolimus/uso terapéutico , Animales , Enfermedad Crónica , Hipertrofia Ventricular Izquierda/prevención & control , Masculino , Nefrectomía , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Remodelación Ventricular/efectos de los fármacos
7.
Kidney Int ; 80(9): 959-969, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21814176

RESUMEN

Podocytes have a significant role in establishing selective permeability of the glomerular filtration barrier. Sustained renin-angiotensin-aldosterone system activation is crucial to the pathogenesis of podocyte injury, but the mechanisms by which angiotensin II modulates podocyte survival due to physiological or injurious stimuli remain unclear. Here, we used proteomic analysis to find new mediators of angiotensin II-induced podocyte injury. Antioxidant protein peroxiredoxin 2 expression was decreased in cultured podocytes stimulated with angiotensin II. Peroxiredoxin 2 was found to be expressed in podocytes in vivo, and its expression was decreased in the glomeruli of rats transgenic for angiotensin II type 1 receptors in a podocyte-specific manner, or in rats infused with angiotensin II. Downregulation of peroxiredoxin 2 in podocytes resulted in increased reactive oxygen species release, protein overoxidation, and inhibition of the Akt pathway. Both treatment with angiotensin II and downregulation of peroxiredoxin 2 expression led to apoptosis of podocytes. Thus, peroxiredoxin 2 is an important modulator of angiotensin II-induced podocyte injury.


Asunto(s)
Angiotensina II/metabolismo , Apoptosis , Glomérulos Renales/enzimología , Peroxirredoxinas/metabolismo , Podocitos/enzimología , Angiotensina II/administración & dosificación , Animales , Línea Celular , Regulación hacia Abajo , Electroforesis en Gel Bidimensional , Humanos , Infusiones Subcutáneas , Glomérulos Renales/patología , Ratones , Estrés Oxidativo , Fosforilación , Podocitos/patología , Proteómica/métodos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Transgénicas , Especies Reactivas de Oxígeno/metabolismo , Receptor de Angiotensina Tipo 1/genética , Receptor de Angiotensina Tipo 1/metabolismo , Transducción de Señal , Factores de Tiempo , Transfección
8.
J Am Soc Nephrol ; 20(10): 2235-45, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19608702

RESUMEN

Endothelial dysfunction contributes to the increased cardiovascular risk that accompanies CKD. We hypothesized that the soluble VEGF receptor 1 (sFlt-1), a VEGF antagonist, plays a role in endothelial dysfunction and decreased angiogenesis in CKD. We enrolled 130 patients with CKD stages 3 to 5 and 56 age- and gender-matched control patients. Plasma sFlt-1 levels were higher in patients with CKD and, after multivariate regression analyses, exclusively associated with renal function and levels of vWF, a marker of endothelial dysfunction. Compared with serum from control patients, both recombinant sFlt-1 and serum from patients with CKD had antiangiogenic activity in the chick chorioallantoic membrane (CAM) assay, induced endothelial cell apoptosis in vitro, and decreased nitric oxide generation in two different endothelial cell lines. Pretreating the sera with an antibody against sFlt-1 abrogated all of these effects. Furthermore, we observed increased sFlt1 levels in 5/6-nephrectomized rats compared with sham-operated animals. Finally, using real-time PCR and ELISA, we identified monocytes as a possible source of increased sFlt-1 in patients with CKD. Our findings show that excess sFlt-1 associates with endothelial dysfunction in CKD and suggest that increased sFlt-1 may predict cardiovascular risk in CKD.


Asunto(s)
Endotelio Vascular/fisiopatología , Enfermedades Renales/fisiopatología , Receptor 1 de Factores de Crecimiento Endotelial Vascular/fisiología , Adulto , Anciano , Apoptosis , Enfermedad Crónica , Células Endoteliales/fisiología , Femenino , Tasa de Filtración Glomerular , Humanos , Masculino , Persona de Mediana Edad , Monocitos/química , Óxido Nítrico/biosíntesis , Receptor 1 de Factores de Crecimiento Endotelial Vascular/sangre
9.
Mol Med ; 14(11-12): 705-14, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18670619

RESUMEN

Hyperglycemia induces defects in angiogenesis without alteration in the expression of major vascular growth factors in the chicken chorioallantoic membrane (CAM) model. A direct negative effect of hyperglycemia on angiogenesis may participate in failures of "therapeutic angiogenesis" trials. Here, we tested the hypothesis that the response to pro-angiogenic molecules such as angiotensin-converting enzyme (ACE), endothelin-1 (ET-1), and vascular endothelial growth factor-A (VEGF) is altered by hyperglycemia. Transfected (Chinese hamster ovary [CHO] or human embryonic kidney [HEK]) cells overexpressing ACE, ET-1, or VEGF were deposed onto the CAM of hyperglycemic or control embryos. The proangiogenic effect was evaluated 3 d later by angiography and histological analyses. Gene expression in response to these factors was assessed by in situ hybridization. Only VEGF overexpression evoked a proangiogenic response in the CAM from hyperglycemic embryos, upregulating the expression of endogenous VEGF, VEGF-R2, and Tie-2, all of them related to activation of endothelial cells. In conclusion, in a model where hyperglycemia does not alter the major vascular growth factor expression, the negative effect of diabetes on capillary density was overcome only by VEGF overexpression, whereas responses to other vasoactive peptides were practically abolished under hyperglycemic conditions.


Asunto(s)
Inductores de la Angiogénesis/metabolismo , Membrana Corioalantoides/metabolismo , Diabetes Mellitus/metabolismo , Angiografía , Animales , Células CHO , Línea Celular , Embrión de Pollo , Pollos , Cricetinae , Cricetulus , Diabetes Mellitus/patología , Endotelina-1/genética , Endotelina-1/fisiología , Humanos , Hiperglucemia/inducido químicamente , Hiperglucemia/fisiopatología , Inmunohistoquímica , Hibridación in Situ , Ratones , Neovascularización Fisiológica , Peptidil-Dipeptidasa A/genética , Peptidil-Dipeptidasa A/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/fisiología
10.
Sci Rep ; 7(1): 9444, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28842629

RESUMEN

The reduced number of circulating stem/progenitor cells that is found in chronic kidney disease (CKD) patients may contribute to impaired angiogenic repair and decreased capillary density in the heart. Cell therapy with bone marrow-derived cells (BMDCs) has been shown to induce positive effects on the microvasculature and cardiac function, most likely due to secretion of growth factors and cytokines, all of which are present in the conditioned medium (CM); however, this is controversial. Here we showed that treatment with BMDC or CM restored vascular density and decreased the extent of fibrosis in a rat model of CKD, the 5/6 nephrectomy. Engraftment and differentiation of exogenous BMDCs could not be detected. Yet CM led to the mobilization and infiltration of endogenous circulating cells into the heart. Cell recruitment was facilitated by the local expression of pro-inflammatory factors such as the macrophage chemoattractant protein-1, interleukin-6, and endothelial adhesion molecules. Consistently, in vitro assays showed that CM increased endothelial adhesiveness to circulating cells by upregulating the expression of adhesion molecules, and stimulated angiogenesis/endothelial tube formation. Overall, our results suggest that both treatments exert vasculoprotective effects on the heart of uremic rats by stimulating endogenous repair mechanisms.


Asunto(s)
Células de la Médula Ósea/fisiología , Trasplante de Médula Ósea , Vasos Coronarios/fisiología , Medios de Cultivo Condicionados/metabolismo , Riñón/fisiología , Insuficiencia Renal Crónica/terapia , Uremia/terapia , Animales , Quimiocina CCL2/metabolismo , Modelos Animales de Enfermedad , Humanos , Interleucina-6/metabolismo , Riñón/cirugía , Masculino , Neovascularización Fisiológica , Ratas , Ratas Sprague-Dawley , Insuficiencia Renal Crónica/inmunología , Uremia/inmunología , Remodelación Vascular
11.
PLoS One ; 6(9): e24046, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931640

RESUMEN

BACKGROUND: Kidney transplantation (RTx) leads to amelioration of endothelial function in patients with advanced renal failure. Endothelial progenitor cells (EPCs) may play a key role in this repair process. The aim of this study was to determine the impact of RTx and immunosuppressive therapy on the number of circulating EPCs. METHODS: We analyzed 52 RTx patients (58±13 years; 33 males, mean ± SD) and 16 age- and gender-matched subjects with normal kidney function (57±17; 10 males). RTx patients received a calcineurin inhibitor (CNI)-based (65%) or a CNI-free therapy (35%) and steroids. EPC number was determined by double positive staining for CD133/VEGFR2 and CD34/VEGFR2 by flow cytometry. Stromal cell-derived factor 1 alpha (SDF-1) levels were assessed by ELISA. Experimentally, to dissociate the impact of RTx from the impact of immunosuppressants, we used the 5/6 nephrectomy model. The animals were treated with a CNI-based or a CNI-free therapy, and EPCs (Sca+cKit+) and CD26+ cells were determined by flow cytometry. RESULTS: Compared to controls, circulating number of CD34+/VEGFR2+ and CD133+/VEGFR2+ EPCs increased in RTx patients. There were no correlations between EPC levels and statin, erythropoietin or use of renin angiotensin system blockers in our study. Indeed, multivariate analysis showed that SDF-1--a cytokine responsible for EPC mobilization--is independently associated with the EPC number. 5/6 rats presented decreased EPC counts in comparison to control animals. Immunosuppressive therapy was able to restore normal EPC values in 5/6 rats. These effects on EPC number were associated with reduced number of CD26+ cells, which might be related to consequent accumulation of SDF-1. CONCLUSIONS: We conclude that kidney transplantation and its associated use of immunosuppressive drugs increases the number of circulating EPCs via the manipulation of the CD26/SDF-1 axis. Increased EPC count may be associated to endothelial repair and function in these patients.


Asunto(s)
Células Endoteliales/citología , Trasplante de Riñón/efectos adversos , Células Madre/citología , Animales , Inhibidores de la Calcineurina , Recuento de Células , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Inmunosupresores/farmacología , Masculino , Ratas , Ratas Sprague-Dawley , Células Madre/efectos de los fármacos
12.
J Cardiovasc Pharmacol ; 49(2): 96-9, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17312450

RESUMEN

Rapamycin (RP; rapamune, sirolimus) is a potent inhibitor of vascular smooth muscle cell proliferation and migration. RP was demonstrated to reduce vascular neointimal formation in different animal models of vascular smooth muscle cell proliferation, and clinical use of RP-eluting stents promotes significant reductions in in-stent restenosis rates. However, high costs still preclude the widespread use of these devices. Oral administration of RP associated to bare metal stent delivery has been advocated as an effective and considerably less expensive alternative for restenosis prevention. It is noteworthy that the presence of RP-eluting stents has been associated with reduced endothelial-dependent vasodilation and coronary spasm. In addition, RP has been demonstrated to prevent vasculogenesis. This study evaluated the effects of RP on endothelium-dependent vascular tone and demonstrated that in vitro incubation with high concentrations of RP did not modify either contraction or relaxation of aortic rings. Similar results were obtained after in vivo administration of the drug. These findings suggest that function of adult, non-proliferative rat endothelial cells involved in vascular tone control is not affected by orally administered RP.


Asunto(s)
Inmunosupresores/farmacología , Músculo Liso Vascular/efectos de los fármacos , Sirolimus/farmacología , Animales , Aorta Torácica/efectos de los fármacos , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Técnicas In Vitro , Masculino , Tono Muscular/efectos de los fármacos , Nitroprusiato/farmacología , Ratas , Ratas Wistar , Vasodilatadores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA