Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
J Mol Cell Cardiol ; 121: 124-133, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29981797

RESUMEN

Managing the cardiovascular complications of renal failure is a major therapeutic challenge in clinical practice. Mineralocorticoid Receptor (MR) blockade is a highly effective strategy for the management of heart failure, but the use of MR antagonists (MRA) is limited by their side effects rendering them contraindicated in patients with renal failure. Finerenone is a new non-steroidal MRA that shows fewer hyperkaliaemic events than the traditional steroidal MRAs and could therefore represent an alternative to these molecules in patients with damaged kidney function. The aim of this study is to characterize the effects of Finerenone on the cardiac complications of renal failure in a mouse model of chronic kidney disease (CKD). CKD was induced by subtotal nephrectomy (Nx), and finerenone was administered at a low dose (2.5 mg/kg/d) from week 4 to week 10 post-Nx. Cardiac function was assessed by echocardiography and invasive hemodynamics while cardiac fibrosis was measured by Sirius Red staining. Renal failure induced cardiac systolic and diastolic dysfunctions in the untreated CKD mice, as well as minor changes on cardiac structure. We also observed alterations in the phosphorylation of proteins playing key roles in the calcium handling (Phospholamban, Calmodulin kinase II) in these mice. Finerenone prevented most of these lesions with no effects on neither the renal dysfunction nor kaliemia. The benefits of finerenone suggest that activation of MR is involved in the cardiac complication of renal failure and strengthen previous studies showing beneficial effects of MRA in patients with CKD.


Asunto(s)
Insuficiencia Cardíaca Diastólica/tratamiento farmacológico , Naftiridinas/administración & dosificación , Receptores de Mineralocorticoides/genética , Insuficiencia Renal Crónica/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Eplerenona/administración & dosificación , Insuficiencia Cardíaca Diastólica/etiología , Insuficiencia Cardíaca Diastólica/genética , Insuficiencia Cardíaca Diastólica/fisiopatología , Hemodinámica/efectos de los fármacos , Humanos , Ratones , Antagonistas de Receptores de Mineralocorticoides/administración & dosificación , Insuficiencia Renal Crónica/complicaciones , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/fisiopatología
2.
Hypertension ; 69(5): 870-878, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28320854

RESUMEN

Acute kidney injury induced by ischemia/reperfusion (IR) is a frequent complication in hospitalized patients. Mineralocorticoid receptor antagonism has shown to be helpful against renal IR consequences; however, the potential benefit of novel nonsteroidal mineralocorticoid receptor antagonists such as finerenone has to be further explored. In this study, we evaluated the efficacy of finerenone to prevent the acute and chronic consequences of ischemic acute kidney injury. For the acute study (24 hours), 18 rats were divided into sham, bilateral renal ischemia of 25 minutes, and rats that received 3 doses of finerenone at 48, 24, and 1 hour before the ischemia. For the chronic study (4 months), 23 rats were divided into sham, rats that underwent 45 minutes of bilateral ischemia, and rats treated with finerenone at days 2 and 1 and 1 hour before IR. We found that after 24 hours of reperfusion, the untreated IR rats presented kidney dysfunction and tubular injury. Kidney injury molecule-1 and neutrophil gelatinase associated to lipolacin mRNA levels were increased. In contrast, the rats treated with finerenone displayed normal kidney function and significantly lesser tubular injury and kidney injury molecule-1 and neutrophil gelatinase associated to lipolacin levels. After 4 months, the IR rats developed chronic kidney disease, evidenced by kidney dysfunction, increased proteinuria and renal vascular resistance, tubular dilation, extensive tubule-interstitial fibrosis, and an increase in kidney transforming growth factor-ß and collagen-I mRNA. The transition from acute kidney injury to chronic kidney disease was fully prevented by finerenone. Altogether, our data show that in the rat, finerenone is able to prevent acute kidney injury induced by IR and the chronic and progressive deterioration of kidney function and structure.


Asunto(s)
Lesión Renal Aguda/metabolismo , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Naftiridinas/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Insuficiencia Renal Crónica/prevención & control , Daño por Reperfusión/metabolismo , Lesión Renal Aguda/complicaciones , Animales , Moléculas de Adhesión Celular/metabolismo , Modelos Animales de Enfermedad , Riñón/efectos de los fármacos , Riñón/metabolismo , Lipocalina 2/metabolismo , Masculino , Malondialdehído/metabolismo , Antagonistas de Receptores de Mineralocorticoides/farmacología , Naftiridinas/farmacología , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/etiología , Insuficiencia Renal Crónica/metabolismo , Daño por Reperfusión/complicaciones
3.
Clin Sci (Lond) ; 130(5): 365-76, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26608078

RESUMEN

ERß (oestrogen receptor ß) activation has been shown to be cardioprotective, but the cell types and mechanisms involved are not understood. To investigate whether ERß restricted to cardiomyocytes contributes to the observed cardioprotection, we tested the effects of cardiomyocyte-specific ERß-OE (ERß overexpression) on survival, cardiac remodelling and function after MI (myocardial infarction) and studied the molecular pathways potentially involved. Female and male mice with cardiomyocyte-specific ERß-OE and WT (wild-type) littermates were subjected to chronic anterior coronary artery ligation or sham surgery. Two weeks after MI, ERß-OE mice showed improved survival (100% and 83% compared with 76% and 58% in WT females and males respectively). ERß-OE was associated with attenuated LV (left ventricular) dilatation, smaller increase in heart weight, less lung congestion at similar MI size, and improved systolic and diastolic function in both sexes. We identified two potential pathways for ERß-mediated myocardial protection. First, male and female ERß-OE mice had a lower reduction of SERCA2a (sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2a) expression after MI, suggesting less reduction in diastolic Ca(2+)-reuptake into the sarcoplasmic reticulum post-MI. Secondly, male ERß-OE revealed attenuated cardiac fibrosis in the remote LV tissue and expression of fibrosis markers collagen I and III, periostin and miR-21. Cardiomyocyte-specific ERß-OE improved survival associated with reduced maladaptive remodelling, improved cardiac function and less heart failure development after MI in both sexes. These effects seem to be related, at least in part, to a better maintenance of Ca(2+) cycling in both sexes and a lower induction of cardiac fibrosis in males after MI.


Asunto(s)
Receptor beta de Estrógeno/metabolismo , Infarto del Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Animales , Biomarcadores/metabolismo , Diástole/fisiología , Receptor beta de Estrógeno/fisiología , Femenino , Fibrosis , Masculino , Ratones Transgénicos , Infarto del Miocardio/diagnóstico por imagen , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocardio/patología , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo , Factores Sexuales , Sístole/fisiología , Ultrasonografía , Remodelación Ventricular/fisiología
4.
Nat Commun ; 6: 8835, 2015 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-26617239

RESUMEN

Despite its high prevalence and economic burden, the aetiology of human hypertension remains incompletely understood. Here we identify the transcription factor GATA5, as a new regulator of blood pressure (BP). GATA5 is expressed in microvascular endothelial cells and its genetic inactivation in mice (Gata5-null) leads to vascular endothelial dysfunction and hypertension. Endothelial-specific inactivation of Gata5 mimics the hypertensive phenotype of the Gata5-null mice, suggestive of an important role for GATA5 in endothelial homeostasis. Transcriptomic analysis of human microvascular endothelial cells with GATA5 knockdown reveals that GATA5 affects several genes and pathways critical for proper endothelial function, such as PKA and nitric oxide pathways. Consistent with a role in human hypertension, we report genetic association of variants at the GATA5 locus with hypertension traits in two large independent cohorts. Our results unveil an unsuspected link between GATA5 and a prominent human condition, and provide a new animal model for hypertension.


Asunto(s)
Presión Sanguínea , Células Endoteliales/metabolismo , Factor de Transcripción GATA5/metabolismo , Hipertensión/metabolismo , Hipertensión/fisiopatología , Animales , Modelos Animales de Enfermedad , Femenino , Factor de Transcripción GATA5/genética , Humanos , Hipertensión/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
5.
J Am Heart Assoc ; 4(1): e001266, 2015 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-25564371

RESUMEN

BACKGROUND: Experimentally, aldosterone in association with NaCl induces cardiac fibrosis, oxidative stress, and inflammation through mineralocorticoid receptor activation; however, the biological processes regulated by aldosterone alone in the heart remain to be identified. METHODS AND RESULTS: Mice were treated for 7 days with aldosterone, and then cardiac transcriptome was analyzed. Aldosterone regulated 60 transcripts (51 upregulated and 9 downregulated) in the heart (fold change ≥1.5, false discovery rate <0.01). To identify the biological processes modulated by aldosterone, a gene ontology analysis was performed. The majority of aldosterone-regulated genes were involved in cell division. The cardiac Ki-67 index (an index of proliferation) of aldosterone-treated mice was higher than that of nontreated mice, confirming microarray predictions. Costaining of Ki-67 with vinculin, CD68, α-smooth muscle actin, CD31, or caveolin 1 revealed that the cycling cells were essentially endothelial cells. Aldosterone-induced mineralocorticoid receptor-dependent proliferation was confirmed ex vivo in human endothelial cells. Moreover, pharmacological-specific blockade of mineralocorticoid receptor by eplerenone inhibited endothelial cell proliferation in a preclinical model of heart failure (transverse aortic constriction). CONCLUSIONS: Aldosterone modulates cardiac gene expression and induces the proliferation of cardiac endothelial cells in vivo.


Asunto(s)
Aldosterona/farmacología , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Endoteliales/efectos de los fármacos , Insuficiencia Cardíaca/metabolismo , Análisis de Varianza , Animales , Presión Sanguínea/fisiología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos , Estadísticas no Paramétricas
6.
J Cell Sci Ther ; 5(1): 153, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24977106

RESUMEN

Experimental studies showed that 17ß-estradiol (E2) and activated Estrogen Receptors (ER) protect the heart from ischemic injury. However, the underlying molecular mechanisms are not well understood. To investigate the role of ER-alpha (ERα) in cardiomyocytes in the setting of myocardial ischemia, we generated transgenic mice with cardiomyocyte-specific overexpression of ERα (ERα-OE) and subjected them to Myocardial Infarction (MI). At the basal level, female and male ERα-OE mice showed increased Left Ventricular (LV) mass, LV volume and cardiomyocyte length. Two weeks after MI, LV volume was significantly increased and LV wall thickness decreased in female and male WT-mice and male ERα-OE, but not in female ERα-OE mice. ERα-OE enhanced expression of angiogenesis and lymphangiogenesis markers (Vegf, Lyve-1), and neovascularization in the peri-infarct area in both sexes. However, attenuated level of fibrosis and higher phosphorylation of JNK signaling pathway could be detected only in female ERα-OE after MI. In conclusion, our study indicates that ERα protects female mouse cardiomyocytes from the sequelae of ischemia through induction of neovascularization in a paracrine fashion and impaired fibrosis, which together may contribute to the attenuation of cardiac remodelling.

7.
PLoS One ; 8(9): e73737, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24040049

RESUMEN

Aldosterone binds to the mineralocorticoid receptor (MR) and exerts pleiotropic effects beyond enhancing renal sodium reabsorption. Excessive mineralocorticoid signaling is deleterious during the evolution of cardiac failure, as evidenced by the benefits provided by adding MR antagonists (MRA) to standard care in humans. In animal models of cardiovascular diseases, MRA reduce cardiac fibrosis. Interestingly diuretics such as torasemide also appear efficient to improve cardiovascular morbidity and mortality, through several mechanisms. Among them, it has been suggested that torasemide could block aldosterone binding to the MR. To evaluate whether torasemide acts as a MRA in cardiomyocytes, we compared its effects with a classic MRA such as spironolactone. We monitored ligand-induced nuclear translocation of MR-GFP and MR transactivation activity in the cardiac-like cell line H9C2 using a reporter gene assay and known endogenous aldosterone-regulated cardiac genes. Torasemide did not modify MR nuclear translocation. Aldosterone-induced MR transactivation activity was reduced by the MRA spironolactone, not by torasemide. Spironolactone blocked the induction by aldosterone of endogenous MR-responsive genes (Sgk-1, PAI-1, Orosomucoid-1, Rgs-2, Serpina-3, Tenascin-X), while torasemide was ineffective. These results show that torasemide is not an MR antagonist; its association with MRA in heart failure may however be beneficial, through actions on complementary pathways.


Asunto(s)
Aldosterona/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Mineralocorticoides/metabolismo , Sulfonamidas/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Animales , Células COS , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Chlorocebus aethiops , Diuréticos/farmacología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Microscopía Confocal , Antagonistas de Receptores de Mineralocorticoides/farmacología , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Orosomucoide/genética , Inhibidor 1 de Activador Plasminogénico/genética , Proteínas Serina-Treonina Quinasas/genética , Ratas , Receptores de Mineralocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Serpinas/genética , Espironolactona/farmacología , Tenascina/genética , Torasemida , Activación Transcripcional/efectos de los fármacos
8.
Hypertension ; 61(2): 361-7, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23297371

RESUMEN

Inappropriate mineralocorticoid receptor (MR) activation is involved in cardiac diseases. Whether and how aldosterone is involved in the deleterious effects of cardiac mineralocorticoid activation is still unclear. Mice overexpressing MR in cardiomyocytes and their controls were treated for 7 days with aldosterone, and cardiac transcriptome was analyzed. Aldosterone regulated 265 genes in cardiomyocyte-targeted MR overexpression mice. Forty three of these genes were also differentially expressed between untreated cardiomyocyte-targeted MR overexpression and controls mice, thus representing putative aldosterone-regulated genes in cardiomyocytes. Among these genes, we focused on connective tissue growth factor (CTGF). In vivo, in cardiomyocyte-targeted MR overexpression mice, aldosterone (but not corticosterone) induced CTGF expression (mRNA and protein) in cardiomyocytes. Ex vivo, aldosterone induced the binding of mineralocorticoid receptor to CTGF promoter and increased the expression of its transcript. Aldosterone induction of CTGF synthesis in cardiomyocytes seems pathologically relevant as the increase in CTGF observed in a model of heart failure (transverse aortic constriction) in rats was prevented by eplerenone, a mineralocorticoid receptor blocker. This study demonstrates that aldosterone specifically regulates gene expression in cardiomyocytes despite large prevalence of glucocorticoids in plasma.


Asunto(s)
Aldosterona/farmacología , Miocitos Cardíacos/efectos de los fármacos , Receptores de Mineralocorticoides/metabolismo , Transcriptoma/efectos de los fármacos , Animales , Factor de Crecimiento del Tejido Conjuntivo/genética , Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Eplerenona , Ratones , Ratones Transgénicos , Antagonistas de Receptores de Mineralocorticoides/farmacología , Miocitos Cardíacos/metabolismo , Receptores de Mineralocorticoides/genética , Espironolactona/análogos & derivados , Espironolactona/farmacología
9.
Hypertension ; 59(5): 966-72, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22469622

RESUMEN

Mineralocorticoid receptor (MR) activation may be deleterious to the cardiovascular system, and MR antagonists improve morbidity and mortality of patients with heart failure. However, mineralocorticoid signaling in the heart remains largely unknown. Using a pan-genomic transcriptomic analysis, we identified neutrophil gelatinase-associated lipocalin (NGAL or lipocalin 2) as a strongly induced gene in the heart of mice with conditional and targeted MR overexpression in cardiomyocytes (whereas induction was low in glucocorticoid receptor-overexpressing mice). NGAL mRNA levels were enhanced after hormonal stimulation by the MR ligand aldosterone in cultured cardiac cells and in the heart of wild-type mice. Mineralocorticoid pathological challenge induced by nephrectomy/aldosterone/salt treatment upregulated NGAL expression in the heart and aorta and its plasma levels. We show evidence for MR binding to an NGAL promoter, providing a mechanism for NGAL regulation. We propose that NGAL may be a marker of mineralocorticoid-dependent injury in the cardiovascular system in mice.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Lipocalinas/metabolismo , Miocitos Cardíacos/metabolismo , Proteínas Oncogénicas/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/fisiología , Proteínas de Fase Aguda/genética , Análisis de Varianza , Animales , Western Blotting , Sistema Cardiovascular/metabolismo , Células Cultivadas , Modelos Animales de Enfermedad , Humanos , Lipocalina 2 , Lipocalinas/genética , Ratones , Ratones Transgénicos , Miocitos Cardíacos/efectos de los fármacos , Proteínas Oncogénicas/genética , ARN Mensajero/análisis , Distribución Aleatoria , Receptores de Mineralocorticoides/genética , Valores de Referencia , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sensibilidad y Especificidad , Transducción de Señal/genética , Regulación hacia Arriba
10.
PLoS One ; 7(1): e30156, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22291909

RESUMEN

Experimental and clinical studies have shown that aldosterone/mineralocorticoid receptor (MR) activation has deleterious effects in the cardiovascular system; however, the signalling pathways involved in the pathophysiological effects of aldosterone/MR in vivo are not fully understood. Several in vitro studies suggest that Epidermal Growth Factor Receptor (EGFR) plays a role in the cardiovascular effects of aldosterone. This hypothesis remains to be demonstrated in vivo. To investigate this question, we analyzed the molecular and functional consequences of aldosterone exposure in a transgenic mouse model with constitutive cardiomyocyte-specific overexpression of a mutant EGFR acting as a dominant negative protein (DN-EGFR). As previously reported, Angiotensin II-mediated cardiac remodelling was prevented in DN-EGFR mice. However, when chronic MR activation was induced by aldosterone-salt-uninephrectomy, cardiac hypertrophy was similar between control littermates and DN-EGFR. In the same way, mRNA expression of markers of cardiac remodelling such as ANF, BNF or ß-Myosin Heavy Chain as well as Collagen 1a and 3a was similarly induced in DN-EGFR mice and their CT littermates. Our findings confirm the role of EGFR in AngII mediated cardiac hypertrophy, and highlight that EGFR is not involved in vivo in the damaging effects of aldosterone on cardiac function and remodelling.


Asunto(s)
Aldosterona/farmacología , Angiotensina II/farmacología , Receptores ErbB/fisiología , Cloruro de Sodio Dietético/farmacología , Remodelación Ventricular/efectos de los fármacos , Remodelación Ventricular/genética , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Corazón/efectos de los fármacos , Corazón/fisiología , Masculino , Ratones , Ratones Transgénicos , Nefrectomía , Sales (Química)/efectos adversos , Sales (Química)/farmacología , Cloruro de Sodio Dietético/efectos adversos , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Remodelación Ventricular/fisiología
11.
Mol Cell Endocrinol ; 350(2): 266-72, 2012 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-21784127

RESUMEN

Several large clinical studies have demonstrated the important benefit of mineralocorticoid receptor (MR) antagonists in patients with heart failure, left ventricular dysfunction after myocardial infarction, hypertension or diabetic nephropathy. Aldosterone adjusts the hydro-mineral balance in the body, and thus participates decisively to the control of blood pressure. This traditional view of the action of aldosterone restricted to sodium reabsorption in epithelial tissues must be revisited. Clinical and experimental studies indicated that chronic activation of the MR in target tissues induces structural and functional changes in the heart, kidneys and blood vessels. These deleterious effects include cardiac and renal fibrosis, inflammation and vascular remodeling. It is important to underscore that these effects are due to elevated MR activation that is inadequate for the body salt requirements. Aldosterone is generally considered as the main ligand of MR. However, this is a matter of debate especially in heart. Complexity arises from the glucocorticoids with circulating concentrations much higher than those of aldosterone, and the fact that the MR has a high affinity for 11ß-hydroxyglucocorticoids. Nevertheless, the beneficial effects of MR inhibition in patients with heart failure emphasize the importance of this receptor in cardiovascular tissue. Diverse experimental models and strains of transgenic mice have allowed to dissect the effects of aldosterone and the MR in the heart. Taken together experimental and clinical data clearly highlight the deleterious cardiovascular effects of MR stimulation.


Asunto(s)
Aldosterona/fisiología , Insuficiencia Cardíaca/etiología , Receptores de Mineralocorticoides/fisiología , Aldosterona/metabolismo , Animales , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Hidrocortisona/metabolismo , Hidrocortisona/fisiología , Ratones , Antagonistas de Receptores de Mineralocorticoides/farmacología , Receptores de Mineralocorticoides/genética , Receptores de Mineralocorticoides/metabolismo
12.
Hypertension ; 57(2): 238-44, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21199998

RESUMEN

Pathophysiological aldosterone (aldo)/mineralocorticoid receptor signaling has a major impact on the cardiovascular system, resulting in hypertension and vascular remodeling. Mineralocorticoids induce endothelial dysfunction, decreasing vasorelaxation in response to acetylcholine and increasing the response to vasoconstrictors. Activation of the epidermal growth factor receptor (EGFR) is thought to mediate the vascular effects of aldo, but this has yet to be demonstrated in vivo. In this study, we analyzed the molecular and functional vascular consequences of aldo-salt challenge in the waved 2 mouse, a genetic model with a partial loss of EGFR tyrosine kinase activity. Deficient EGFR activity is associated with global oxidative stress and endothelial dysfunction. A decrease in EGFR activity did not affect the arterial wall remodeling process induced by aldo-salt. By contrast, normal EGFR activity was required for the aldo-induced enhancement of phenylephrine- and angiotensin II-mediated vasoconstriction. In conclusion, this in vivo study demonstrates that EGFR plays a key role in aldosterone-mediated vascular reactivity.


Asunto(s)
Aldosterona/farmacología , Aorta/efectos de los fármacos , Receptores ErbB/fisiología , Vasoconstricción/efectos de los fármacos , Acetilcolina/farmacología , Angiotensina II/farmacología , Animales , Aorta/metabolismo , Aorta/fisiopatología , Western Blotting , Arterias Carótidas/efectos de los fármacos , Arterias Carótidas/fisiopatología , Relación Dosis-Respuesta a Droga , Receptores ErbB/genética , Receptores ErbB/metabolismo , Expresión Génica/efectos de los fármacos , Genotipo , Hemodinámica/efectos de los fármacos , Técnicas In Vitro , Masculino , Ratones , Ratones Mutantes , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Nefrectomía , Óxido Nítrico Sintasa de Tipo III/metabolismo , Nitroprusiato/farmacología , Fenilefrina/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Vasoconstrictores/farmacología , Vasodilatadores/farmacología
14.
FASEB J ; 23(7): 2176-85, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19255254

RESUMEN

Recent reports showed an unexpected worsening of endothelial function by aldosterone antagonism in diabetic patients, suggesting that aldosterone could interfere with the detrimental consequences of diabetes on microvasculature and thus on cardiac function. To test this hypothesis, diabetes (D) was induced using streptozotocin in transgenic (Tg) male mice overexpressing aldosterone-synthase in the heart and in wild-type (Wt) mice. Eight weeks after streptozotocin injection, impairment of left ventricular systolic function, measured by echocardiography (fractional shortening), was accompanied by a decrease in capillaries/cardiomyocyte ratio (-20%) and VEGFa expression (-40%) in Wt-D mice compared with normoglycemic littermates. Furthermore, Wt-D mice demonstrated an increase in superoxide production (+100%) and protein carbonylation (+33%), hallmarks of oxidative stress. Except for a slight increase in protein carbonylation, all of these diabetes-associated cardiac alterations were undetectable in Tg-D mice. Fibrosis was induced similarly in both diabetic groups. Eplerenone (an aldosterone antagonist) abolished all of the effects of aldosterone-synthase overexpression but had no effect in Wt-D mice. Thus, aldosterone prevents systolic dysfunction through a mineralocorticoid receptor-dependent mechanism that may include preventing VEGFa down-regulation and maintaining capillary density. Understanding how aldosterone prevents VEGFa down-regulation in experimental diabetes could be important to define new strategies targeting the prevention of a decrease in capillary density.


Asunto(s)
Aldosterona/fisiología , Capilares , Vasos Coronarios , Regulación de la Expresión Génica/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Aldosterona/genética , Animales , Complicaciones de la Diabetes , Diabetes Mellitus Experimental , Corazón , Ratones , Miocardio , Receptores de Mineralocorticoides/fisiología , Sístole , Disfunción Ventricular Izquierda
15.
Pharmacol Rep ; 61(1): 58-66, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19307693

RESUMEN

Our understanding of the effects of aldosterone and its mechanisms has increased substantially in recent years, probably because of the importance of the mineralocorticoid receptor (MR) antagonists in several major cardiovascular diseases. Recent clinical studies have confirmed the benefits of MR antagonists in patients with heart failure, left ventricular dysfunction after myocardial infarction, hypertension or diabetic nephropathy. However, it would be a gross oversimplification to conclude that the role of aldosterone is unequivocally negative. Aldosterone is synthesized in the adrenal glands and binds to specific MRs in target epithelial cells. The steroid-receptor complex penetrates the cell nucleus where it modulates gene expression and activates specific aldosterone-induced proteins that control sodium reabsorption. Recent studies have shown that aldosterone also impacts a wide range of non-epithelial tissues such as the heart and blood vessels. Remarkably, aldosterone can also be synthesized in extra-adrenal tissues and it may act in a rapid non-genomic manner.We note the existence of glucocorticoids that exhibit plasma concentrations much higher than those of aldosterone and that are structurally very similar to aldosterone. It is thus possible that glucocorticoids may bind to the aldosterone receptor in some cell types. Diverse experimental models and several strains of transgenic mice have allowed us to better understand the effects of aldosterone on the heart. Specifically, it seems that a slight increase in cardiac aldosterone concentrations induces a decreased coronary reserve in mice by decreasing the BKCa potassium channels associated with coronary smooth muscle cells. Taken together, these experiments indicate that vascular cells are the primary targets of aldosterone in the cardiovascular system. The hormone directly affects NO and EDHF-mediated coronary relaxation. Both mechanisms may contribute to the deleterious cardiovascular effects of MR stimulation.


Asunto(s)
Aldosterona/metabolismo , Enfermedades Cardiovasculares/fisiopatología , Antagonistas de Receptores de Mineralocorticoides , Aldosterona/biosíntesis , Angiotensina II/metabolismo , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Ensayos Clínicos como Asunto , Humanos , Transducción de Señal
16.
Am J Physiol Heart Circ Physiol ; 296(2): H435-41, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19074674

RESUMEN

We tested the hypothesis that heart rate (HR) reduction, induced by the selective hyperpolarization-activated current inhibitor ivabradine (Iva), might improve left ventricular (LV) function, structure, and electrical remodeling in severe post-myocardial infarction (MI) chronic heart failure (HF). MI was produced in adult male Wistar rats. After 2 mo, echocardiography was performed before the randomization into MI and MI + Iva (10 mg x kg(-1) x day(-1)) groups. After 3 mo of treatment, echocardiography and 24-h telemetry were recorded. Cardiac collagen, mRNA, and protein expressions of angiotensin-converting enzyme (ACE) and ANG II type 1 (AT(1)) receptor were quantified. As a result, at 2 mo post-MI, all rats displayed severe congestive HF signs (ejection fraction < 30%). At 5 mo post-MI, body and heart weights were similar in the MI and MI + Iva groups. LV ejection fraction and LV end-diastolic pressure were worsened in the MI group, whereas both were improved with Iva. Iva reduced HR by 10.4% (P < 0.03 vs. MI) and ventricular premature complexes by 89% (P < 0.03) and improved HR variability (standard deviation of the RR interval) by 22% (P < 0.05). There were no effects of Iva on PR, QRS, and QT durations. Interstitial fibrosis in the MI-remote LV was markedly reduced by Iva (4.0 +/- 0.1 vs. 1.8 +/- 0.1%, P < 0.005). Increases in ventricular gene and protein expressions of ACE and AT(1) receptor in MI were completely blunted by Iva. In conclusion, these data indicated that HR reduction by Iva prevents the worsening of LV dysfunction and remodeling that may be related to a downregulation of cardiac renin-angiotensin-aldosterone system transcripts. Such beneficial effects of Iva on cardiac remodeling open new clinical perspectives for the treatment of severe HF.


Asunto(s)
Antiarrítmicos/administración & dosificación , Benzazepinas/administración & dosificación , Insuficiencia Cardíaca/tratamiento farmacológico , Frecuencia Cardíaca/efectos de los fármacos , Miocardio/patología , Disfunción Ventricular Izquierda/tratamiento farmacológico , Función Ventricular Izquierda/efectos de los fármacos , Remodelación Ventricular/efectos de los fármacos , Animales , Colágeno/metabolismo , Modelos Animales de Enfermedad , Esquema de Medicación , Ecocardiografía Doppler , Electrocardiografía Ambulatoria , Fibrosis , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/patología , Insuficiencia Cardíaca/fisiopatología , Ivabradina , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Peptidil-Dipeptidasa A/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Wistar , Receptor de Angiotensina Tipo 1/metabolismo , Sistema Renina-Angiotensina/efectos de los fármacos , Sistema Renina-Angiotensina/genética , Índice de Severidad de la Enfermedad , Volumen Sistólico/efectos de los fármacos , Telemetría , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología , Presión Ventricular/efectos de los fármacos
17.
Hypertension ; 52(6): 1060-7, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18981328

RESUMEN

Experimental and clinical studies show that aldosterone/mineralocorticoid receptor (MR) activation has deleterious effects in the cardiovascular system that may cross-talk with those of angiotensin II (Ang II). This study, using a transgenic mouse model with conditional and cardiomyocyte-restricted overexpression of the human MR, was designed to assess the cardiac consequences of Ang II treatment and cardiomyocyte MR activation. Two-month-old MHCtTA/tetO-hMR double transgenic males (DTg) with conditional, cardiomyocyte-specific human MR expression, and their control littermates were infused with Ang II (200 ng/kg per minute) or vehicle via osmotic minipump. Ang II induced similar increases in systolic blood pressure in control and DTg mice but a greater increase in left ventricle mass/body weight in DTg than in control mice. In DTg mice, Ang II-induced left ventricle hypertrophy and diastolic dysfunction without affecting systolic function, as assessed by echography. These effects were associated with an increase in the expression of collagens and fibronectin, matrix metalloproteinase 2 and matrix metalloproteinase 9 activities, and histological fibrosis. Ang II treatment of DTg mice did not affect inflammation markers, but oxidative stress was substantially increased, as indicated by gp91 expression, apocynin-inhibitable NADPH oxidase activity, and protein carbonylation. These molecular and functional alterations were prevented by pharmacological MR antagonism. Our findings indicate that the effects of Ang II and MR activation in the heart are additive. This observation may be relevant to the clinical use of MR or of combined Ang II type 1 receptor-MR antagonists for hypertrophic cardiomyopathies or for heart failure, particularly when diastolic dysfunction is associated with preserved systolic function.


Asunto(s)
Angiotensina II/metabolismo , Insuficiencia Cardíaca/metabolismo , Hipertensión/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/fisiología , Remodelación Ventricular/fisiología , Angiotensina II/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Ecocardiografía , Femenino , Insuficiencia Cardíaca/diagnóstico por imagen , Insuficiencia Cardíaca/fisiopatología , Humanos , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Masculino , Ratones , Ratones Transgénicos , Miocitos Cardíacos/metabolismo , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Embarazo , Receptor Cross-Talk , Receptor de Angiotensina Tipo 1/metabolismo , Receptores de Mineralocorticoides/genética , Vasoconstrictores/metabolismo , Vasoconstrictores/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...