Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 85
Filtrar
1.
Pol Arch Intern Med ; 133(10)2023 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-37818947

RESUMEN

Primary aldosteronism (PA), characterized by inappropriately high concentrations of the adrenal-derived hormone aldosterone, is the most common endocrine cause of arterial hypertension. As compared with individuals with essential hypertension, patients with PA have a significantly increased cardiovascular risk that cannot be fully reversed by common antihypertensive treatment because of blood pressure-independent deleterious effects of aldosterone. Measurement of the aldosterone to renin ratio (ARR), reflecting the degree of aldosterone excess, is the classic screening test for PA, but thresholds for an elevated ARR vary substantially and are arbitrary, as there exists a wide disease continuum that spans from preclinical stages to overt PA. Treatment approaches for PA with either mineralocorticoid receptor antagonists for bilateral disease or unilateral adrenalectomy for aldosterone-producing adenomas (APA) are highly effective to mitigate the excess cardiovascular risk associated with PA. Subtype classification according to the dichotomous concept of unilateral PA, mainly due to APAs, vs bilateral PA, mainly due to bilateral adrenal hyperplasia, has been recently challenged by advances in the pathophysiologic understanding and therapeutic spectrum of PA. The implementation of current PA guidelines into clinical routine is extremely poor, as reflected by the fact that most patients suffering from PA remain undiagnosed and probably untreated. Pragmatic approaches are required to address this public health problem. In this review, we present an up­to­date overview on the clinical significance, diagnosis, and treatment of PA, with the aim to provide guidance for clinicians regarding the management of this disease, paying particular attention to its feasible implementation into daily clinical routine.


Asunto(s)
Hiperaldosteronismo , Humanos , Adrenalectomía , Adenoma Corticosuprarrenal/complicaciones , Aldosterona/efectos adversos , Hiperaldosteronismo/diagnóstico , Hiperaldosteronismo/terapia , Hipertensión/diagnóstico , Hipertensión/tratamiento farmacológico , Hipertensión/etiología
2.
Exp Biol Med (Maywood) ; 248(20): 1768-1779, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37828834

RESUMEN

An increase in blood pressure by a high-salt (HS) diet may change the expression levels of renal epithelial sodium channels (ENaCs) and aquaporins (AQPs). Spontaneously hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats were exposed to HS and regular-salt (RS) diets for 6 weeks. Mean arterial pressure (MAP) and plasma atrial natriuretic peptide (ANP), angiotensin II (Ang II), aldosterone, and arginine vasopressin (AVP) levels were determined. Expression of mRNA levels of ENaCs and AQPs were quantified by real-time PCR. The MAP was higher in SHRs on the HS diet. Plasma Ang II and aldosterone levels were low while plasma ANP level was high in both strains of rats. Renal expression of mRNA levels of α-, ß-, and γ-ENaCs was lowered in SHRs on the HS diet. Meanwhile, renal AQP1, AQP2, and AQP7 mRNA expression levels were lowered in both strains of rats on the HS diet. Suppression of mRNA expression levels of ENaC and AQP subunits suggests that the high-salt-induced increase in the MAP of SHR may not be solely due to renal sodium and water retention.


Asunto(s)
Hipertensión , Ratas , Animales , Hipertensión/metabolismo , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Aldosterona/efectos adversos , Acuaporina 2 , Ratas Endogámicas WKY , Presión Sanguínea , Ratas Endogámicas SHR , Cloruro de Sodio Dietético/efectos adversos , Angiotensina II/efectos adversos , Dieta , ARN Mensajero/genética , Factor Natriurético Atrial/metabolismo , Factor Natriurético Atrial/farmacología
3.
Exp Cell Res ; 431(1): 113738, 2023 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-37572787

RESUMEN

Epithelial-mesenchymal transition (EMT) plays a critical role in hypertension-induced renal fibrosis, a final pathway that leads to end-stage renal failure. C-Atrial natriuretic peptide (ANP)4-23, a specific agonist of natriuretic peptide receptor-C (NPR-C), has been reported to have protective effects against hypertension. However, the role of C-ANP4-23 in hypertension-associated renal fibrosis has not yet been elucidated. In this study, mice were randomly divided into SHAM group, DOCA-salt group and DOCA-salt + C-ANP4-23 group. Renal morphology changes, renal function and fibrosis were detected. Human proximal tubular epithelial cells (HK2) stimulated by aldosterone were used for cell function and mechanism study. The DOCA-salt treated mice exhibited hypertension, kidney fibrosis and renal dysfunction, which were attenuated by C-ANP4-23. Moreover, C-ANP4-23 inhibited DOCA-salt treatment-induced renal EMT as evidenced by decrease of the mesenchymal marker alpha-smooth muscle actin (ACTA2) and vimentin and increase of epithelial cell marker E-cadherin. In HK2 cells, aldosterone induced EMT response, which was also suppressed by C-ANP4-23. The key transcription factors (twist, snail, slug and ZEB1) involved in EMT were increased in the kidney of DOCA-salt-treated mice, which were also suppressed by C-ANP4-23. Mechanistically, C-ANP4-23 inhibited the aldosterone-induced translocation of MR from cytosol to nucleus without change of MR expression. Furthermore, C-ANP4-23 rescued the enhanced expression of NADPH oxidase (NOX) 4 and oxidative stress after aldosterone stimulation. Aldosterone-induced Akt and Erk1/2 activation was also suppressed by C-ANP4-23. Our data suggest that C-ANP4-23 attenuates renal fibrosis, likely through inhibition of MR activation, enhanced oxidative stress and Akt and Erk1/2 signaling pathway.


Asunto(s)
Acetato de Desoxicorticosterona , Hipertensión , Enfermedades Renales , Ratones , Humanos , Animales , Factor Natriurético Atrial/genética , Factor Natriurético Atrial/metabolismo , Receptores del Factor Natriurético Atrial/metabolismo , Aldosterona/efectos adversos , Aldosterona/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Acetato de Desoxicorticosterona/efectos adversos , Hipertensión/inducido químicamente , Hipertensión/metabolismo , Riñón/metabolismo , Enfermedades Renales/inducido químicamente , Enfermedades Renales/prevención & control , Acetatos/efectos adversos , Acetatos/metabolismo , Fibrosis
4.
Adv Ther ; 40(11): 4777-4791, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37606716

RESUMEN

INTRODUCTION: Patients receiving cardiorenal-protective renin-angiotensin-aldosterone system inhibitors (RAASis) are at increased risk of developing hyperkalemia, which is associated with increased medical costs. The aim of this study was to evaluate the impact of adding sodium zirconium cyclosilicate (SZC) therapy on 3-month medical costs in patients who experienced hyperkalemia while receiving RAASi therapy. METHODS: The retrospective OPTIMIZE II study used medical and pharmacy claims data from IQVIA PharMetrics® Plus. Patients aged ≥ 18 years who received SZC (≥ 60 day supply over 3 months' follow-up) and continued RAASi between July 2019 and December 2021 (Continue RAASi + SZC cohort) were 1:1 exact and propensity score matched with patients who discontinued RAASi after hyperkalemia diagnosis and did not receive SZC (Discontinue RAASi + no SZC cohort). The primary outcome was hyperkalemia-related medical costs to payers over 3 months; all-cause medical and pharmacy costs were also analyzed. RESULTS: In the Continue RAASi + SZC (n = 467) versus Discontinue RAASi + no SZC (n = 467) cohort, there were significant reductions in mean per-patient hyperkalemia-related medical costs (reduction of $2216.07; p = 0.01) and all-cause medical costs (reduction of $6102.43; p < 0.001); mean hyperkalemia-related inpatient medical costs and all-cause inpatient and emergency department medical costs were significantly reduced. The reduction in all-cause medical cost in the Continue RAASi + SZC cohort offset an increase in the mean per-patient all-cause pharmacy cost (increase of $3117.71; p < 0.001). CONCLUSION: RAASi therapy has well-established cardiorenal benefits. In OPTIMIZE II, management of RAASi-induced hyperkalemia with SZC was associated with lower hyperkalemia-related and all-cause medical costs than RAASi discontinuation without SZC, demonstrating medical cost savings with maintaining RAASi therapy with SZC.


Asunto(s)
Hiperpotasemia , Humanos , Hiperpotasemia/tratamiento farmacológico , Hiperpotasemia/inducido químicamente , Sistema Renina-Angiotensina , Potasio/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Aldosterona/efectos adversos , Estudios Retrospectivos , Antihipertensivos/efectos adversos
5.
Biol Pharm Bull ; 44(8): 1044-1049, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34078775

RESUMEN

Aldosterone induces cardiac electrical and structural remodeling, which leads to the development of heart failure and/or atrial fibrillation (AF). However, it remains unknown whether aldosterone-induced remodeling may modulate the efficacy of anti-AF drugs. In this study, we aimed to jeopardize the structural and functional remodeling by aldosterone in rats with aorto-venocaval shunts (AVS rats) and evaluate the effect of acehytisine in this model. An AVS operation was performed on rats (n = 6, male) and it was accompanied by the intraperitoneal infusion of aldosterone (AVS + Ald) at 2.0 µg/h for 28 d. The cardiopathy was characterized by echocardiography, electrophysiologic and hemodynamic testing, and morphometric examination in comparison with sham-operated rats (n = 3), sham + Ald (n = 6), and AVS (n = 5). Aldosterone accelerated the progression from asymptomatic heart failure to overt heart failure and induced sustained AF resistant to electrical fibrillation in one out of six rats. In addition, it prolonged PR, QT interval and Wenckebach cycle length. Acehytisine failed to suppress AF in the AVS + Ald rats. In conclusion, aldosterone jeopardized electrical remodeling and blunted the electrophysiological response to acehytisine on AF.


Asunto(s)
Aldosterona/efectos adversos , Derivación Arteriovenosa Quirúrgica/efectos adversos , Fibrilación Atrial/etiología , Fármacos Cardiovasculares/farmacología , Fenómenos Electrofisiológicos , Atrios Cardíacos/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Animales , Aorta/cirugía , Remodelación Atrial , Atrios Cardíacos/patología , Atrios Cardíacos/fisiopatología , Masculino , Ratas Wistar , Venas Cavas/cirugía
6.
Cells ; 10(2)2021 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-33671798

RESUMEN

We investigated the role of aldosterone (ALDO) in the development of arterial thrombosis in streptozotocin-induced diabetic rats. To evaluate the effect of endogenous ALDO, the rats underwent adrenalectomy (ADX). ADX reduced the development of arterial thrombosis. A 1 h infusion of ALDO (30 µg/kg/h) enhanced thrombosis in adrenalectomized rats, while this effect was potentiated in diabetic rats. ALDO shortened bleeding time, increased plasma levels of tissue factor (TF) and plasminogen activator inhibitor, decreased plasma level of nitric oxide (NO) metabolites, and increased oxidative stress. Moreover, 2 h incubation of human umbilical vein endothelial cells (HUVECs) with ALDO (10-7 M) disrupted hemostatic balance in endothelial cells in normoglycemia (glucose 5.5 mM), and this effect was more pronounced in hyperglycemia (glucose 30 mM). We demonstrated that the acute ALDO infusion enhances arterial thrombosis in rats and hyperglycemia potentiates this prothrombotic effect. The mechanism of ALDO action was partially mediated by mineralocorticoid (MR) and glucocorticoid (GR) receptors and related to impact of the hormone on primary hemostasis, TF-dependent coagulation cascade, fibrinolysis, NO bioavailability, and oxidative stress balance. Our in vitro study confirmed that ALDO induces prothrombotic phenotype in the endothelium, particularly under hyperglycemic conditions.


Asunto(s)
Aldosterona/efectos adversos , Aldosterona/uso terapéutico , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/etiología , Animales , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Ratas , Ratas Wistar , Trombosis/etiología , Trombosis/fisiopatología
7.
J Hum Hypertens ; 35(7): 588-597, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-32839534

RESUMEN

The aim of this study was to investigate the possible relationship between worse clinical outcomes and the use of angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs) in hospitalized COVID-19 patients. A total of 247 adult patients (154 males, 93 females; mean age: 51.3 ± 14.2 years) hospitalized for COVID-19 as confirmed by polymerase chain reaction (PCR) were retrospectively reviewed. Demographic and clinical characteristics and laboratory parameters were analyzed using various statistical modeling. Primary outcomes were defined as the need for intensive care unit (ICU), mechanical ventilation, or occurrence of death. Of the patients, 48 were treated in the ICU with a high flow oxygen/noninvasive mechanical ventilation (NIMV, n = 12) or mechanical ventilation (n = 36). Median length of ICU stay was 13 (range, 7-18) days. Mortality was seen in four of the ICU patients. Other patients were followed in the COVID-19 services for a median of 7 days. There was no significant correlation between the primary outcomes and use of ACEIs/ARBs (frequentist OR = 0.82, 95% confidence interval (CI) 0.29-2.34, p = 0.715 and Bayesian posterior median OR = 0.80, 95% CI 0.31-2.02) and presence of hypertension (frequentist OR = 1.23, 95% CI 0.52-2.92, p = 0.631 and Bayesian posterior median OR = 1.25, 95% CI 0.58-2.60). Neutrophil-to-lymphocyte ratio (NLR) and D-dimer levels were strongly associated with primary outcomes. In conclusion, the presence of hypertension and use of ACEIs/ARBs were not significantly associated with poor primary clinical outcomes; however, NLR and D-dimer levels were strong predictors of clinical worsening.


Asunto(s)
Antagonistas de Receptores de Angiotensina/uso terapéutico , Inhibidores de la Enzima Convertidora de Angiotensina/uso terapéutico , COVID-19/diagnóstico , Hipertensión/tratamiento farmacológico , SARS-CoV-2/aislamiento & purificación , Adulto , Anciano , Aldosterona/efectos adversos , Aldosterona/uso terapéutico , Antagonistas de Receptores de Angiotensina/efectos adversos , Inhibidores de la Enzima Convertidora de Angiotensina/efectos adversos , Prueba de Ácido Nucleico para COVID-19 , Femenino , Productos de Degradación de Fibrina-Fibrinógeno/análisis , Humanos , Hipertensión/diagnóstico , Linfocitos , Masculino , Persona de Mediana Edad , Neutrófilos , Reacción en Cadena de la Polimerasa , Sistema Renina-Angiotensina , Estudios Retrospectivos , SARS-CoV-2/genética
8.
Can J Physiol Pharmacol ; 98(9): 579-586, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32437627

RESUMEN

Previously, we showed that global knockout (KO) of the circadian clock transcription factor PER1 in male, but not female, mice fed a high-salt diet plus mineralocorticoid treatment (HS/DOCP) resulted in nondipping hypertension and decreased night/day ratio of sodium (Na) excretion. Additionally, we have shown that the endothelin-1 (ET-1) gene is targeted by both PER1 and aldosterone. We hypothesized that ET-1 would exhibit a sex-specific response to HS/DOCP treatment in PER1 KO. Here we show that male, but not female, global PER1 KO mice exhibit a decreased night/day ratio of urinary ET-1. Gene expression analysis revealed significant genotype differences in ET-1 and endothelin A receptor (ETA) expression in male, but not female, mice in response to HS/DOCP. Additionally, both wild-type and global PER1 KO male mice significantly increase endothelin B receptor (ETB) expression in response to HS/DOCP, but female mice do not. Finally, siRNA-mediated knockdown of PER1 in mouse cortical collecting duct cells (mpkCCDc14) resulted in increased ET-1 mRNA expression and peptide secretion in response to aldosterone treatment. These data suggest that PER1 is a negative regulator of ET-1 expression in response to HS/DOCP, revealing a novel mechanism for the regulation of renal Na handling in response to HS/DOCP treatment.


Asunto(s)
Endotelina-1/metabolismo , Hipertensión/metabolismo , Túbulos Renales Colectores/fisiopatología , Proteínas Circadianas Period/metabolismo , Eliminación Renal/fisiología , Aldosterona/administración & dosificación , Aldosterona/efectos adversos , Animales , Relojes Circadianos/fisiología , Modelos Animales de Enfermedad , Endotelina-1/orina , Femenino , Humanos , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , Proteínas Circadianas Period/genética , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Eliminación Renal/efectos de los fármacos , Factores Sexuales , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo
9.
Sci Rep ; 10(1): 553, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31953473

RESUMEN

Atrial fibrillation (AF) is a progressive arrhythmia with underlying mechanisms that are not fully elucidated, partially due to lack of reliable and affordable animal models. Here, we introduce a system for long-term assessment of AF susceptibility (substrate) in ambulatory rats implanted with miniature electrodes on the atrium. Rats were subjected to excessive aldosterone (Aldo) or solvent only (Sham). An additional group was exposed to myocardial infarction (MI). AF substrate was tested two- and four-weeks post implantation and was also compared with implanted rats early post-implantation (Base). Aldo and MI increased the AF substrate and atrial fibrosis. In the MI group only, AF duration was correlated with the level of atrial fibrosis and was inversely correlated with systolic function. Unexpectedly, Shams also developed progressive AF substrate relative to Base individuals. Further studies indicated that serum inflammatory markers (IL-6, TNF-alpha) were not elevated in the shams. In addition, we excluded anxiety\depression due to social-isolation as an AF promoting factor. Finally, enhanced biocompatibility of the atrial electrode did not inhibit the gradual development of AF substrate over a testing period of up to 8 weeks. Overall, we successfully validated the first system for long-term AF substrate testing in ambulatory rats.


Asunto(s)
Aldosterona/efectos adversos , Fibrilación Atrial/patología , Interleucina-6/sangre , Infarto del Miocardio/patología , Factor de Necrosis Tumoral alfa/sangre , Animales , Fibrilación Atrial/inducido químicamente , Fibrilación Atrial/metabolismo , Modelos Animales de Enfermedad , Electrodos Implantados , Fibrosis , Masculino , Microelectrodos , Infarto del Miocardio/sangre , Ratas , Ratas Sprague-Dawley
10.
Curr Hypertens Rep ; 21(12): 94, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31741119

RESUMEN

PURPOSE: Atrial fibrillation is the most common sustained arrhythmia, with a prevalence of 1-2% in the general population and over 15% in people older than 80 years. Due to aging of the population it imposes an increasing burden on the healthcare system because of the need for life-long pharmacological treatment and the associated increased risk of heart failure and hospitalization. Hence, identification of the factors that predispose to atrial fibrillation it is of utmost relevance. RECENT FINDINGS: Several conditions exist that are characterized by inappropriately high levels of aldosterone, mostly primary aldosteronism and the severe or drug-resistant forms of arterial hypertension. In these forms, aldosterone can cause prominent target organ damage, mostly in the heart, vasculature, and kidney. This review examines the experimental data and clinical evidences that support a link between hyperaldosteronism and atrial fibrillation, and how this knowledge should lead to a change in our management of the hypertensive patients presenting with atrial fibrillation.


Asunto(s)
Aldosterona/sangre , Fibrilación Atrial/fisiopatología , Hiperaldosteronismo/fisiopatología , Hipertensión/fisiopatología , Aldosterona/efectos adversos , Aldosterona/fisiología , Fibrilación Atrial/sangre , Fibrilación Atrial/etiología , Insuficiencia Cardíaca/sangre , Insuficiencia Cardíaca/etiología , Insuficiencia Cardíaca/fisiopatología , Humanos , Hiperaldosteronismo/sangre , Hiperaldosteronismo/complicaciones , Hipertensión/sangre , Hipertensión/etiología , Ensayos Clínicos Controlados Aleatorios como Asunto
11.
Intern Med ; 58(4): 497-504, 2019 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-30333389

RESUMEN

Objective The association of primary aldosteronism (PA) with thyroid disease has already been suggested. The aim of this study was to examine the presence of PA in patients with papillary thyroid carcinoma (PC) and to characterize such PC patients with PA. Methods We examined the presence of PA in 81 consecutive patients with PC, whose random sitting blood pressure (BP) was ≥140/90 mmHg in the office (n= 68), who had an incidental adrenal tumor or adrenal enlargement (n=9), or who showed hypokalemia (n=4). Thirty-one of these 81 patients had been treated with anti-hypertensive drugs. The plasma aldosterone concentration (PAC) and plasma renin activity (PRA) were first measured before operation in 16 patients and after operation in 65 patients. PA was diagnosed according to the guidelines of the Japan Endocrine Society. Results Forty patients with PC with a random PAC/PRA ratio of over 200 were subjected to a further study (12 of these patients had been treated with anti-hypertensive drugs). Ultimately, 15 patients with PC were diagnosed with PA. Adrenal venous sampling was done in 9 out of 15 patients with PC associated with PA. No patients were diagnosed as having unilateral lesions. Among the 15 patients, white-coat hypertension was observed in 5 patients, and normotension was observed in 1 patient. Conclusion These findings suggest that the prevalence of PA may be high among patients with PC. An active examination is needed to detect PA, as its signs and symptoms may be mild in patients with PC associated with hypertension.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/sangre , Aldosterona/efectos adversos , Aldosterona/sangre , Hiperaldosteronismo/sangre , Hiperaldosteronismo/complicaciones , Cáncer Papilar Tiroideo/sangre , Neoplasias de la Tiroides/sangre , Neoplasias de las Glándulas Suprarrenales/complicaciones , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Femenino , Humanos , Hiperaldosteronismo/fisiopatología , Japón , Masculino , Persona de Mediana Edad , Prevalencia , Renina/sangre , Cáncer Papilar Tiroideo/etiología , Cáncer Papilar Tiroideo/fisiopatología , Neoplasias de la Tiroides/etiología , Neoplasias de la Tiroides/fisiopatología
12.
Invest Ophthalmol Vis Sci ; 59(8): 3355-3365, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-30025072

RESUMEN

Purpose: To determine the effects of aldosterone exposure on retinal edema and retinopathy in a mouse model of retinal vein occlusion (RVO). Methods: RVO was induced immediately following intravenous injection of Rose bengal (66 mg/kg) using a 532-nm wavelength laser to place three to seven applications at 80 mW and 50-µm spot size directed at the superior retinal vein one disc diameter away from the nerve. Negative control consisted of placing an equal number of laser spots without targeting the vein. Male and female C57BL/6J mice aged 7 to 9 months with confirmed absence of Crb1rd8 were used. Aldosterone pellets releasing a daily dose of 0.83 µg/day were implanted subcutaneously 4 weeks prior to RVO. Retinal imaging by optical coherence tomography (OCT) was performed using a Micron IV rodent imaging system. Retinas were analyzed by immunohistochemistry using standard techniques. Retinal imaging and tissue analysis were performed 2, 4, and 7 days following RVO. Comparisons were made using Student's t-test, ANOVA, and Pearson's χ2. Results: RVO caused retinal edema in the form of cystic spaces and retinal thickening detectable by both OCT and histology. RVO also caused Müller glia (MG) dysfunction manifest as upregulated glial fibrillary acidic protein (GFAP) and altered localization of aquaporin 4 (AQP4) and Kir4.1. Treatment with aldosterone caused a significant increase in retinal edema and more severe retinopathy manifest as retinal whitening and extensive intraretinal hemorrhage. MG dysfunction was more severe and persistent in aldosterone-treated mice. Finally, aldosterone greatly increased the number of infiltrating mononuclear phagocytes following RVO. Conclusions: Systemic aldosterone exposure causes a more severe RVO phenotype manifest as increased severity and duration of retinal edema and more severe retinopathy. The effects of aldosterone may be mediated by MG dysfunction and increased infiltration of mononuclear phagocytes. This suggests that small increases in aldosterone levels may be a risk factor for severe RVO.


Asunto(s)
Aldosterona/efectos adversos , Modelos Animales de Enfermedad , Coagulación con Láser/efectos adversos , Edema Macular/inducido químicamente , Hemorragia Retiniana/inducido químicamente , Oclusión de la Vena Retiniana/etiología , Aldosterona/administración & dosificación , Animales , Biomarcadores/metabolismo , Western Blotting , Implantes de Medicamentos , Femenino , Colorantes Fluorescentes , Edema Macular/metabolismo , Edema Macular/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Hemorragia Retiniana/metabolismo , Hemorragia Retiniana/fisiopatología , Oclusión de la Vena Retiniana/metabolismo , Oclusión de la Vena Retiniana/fisiopatología , Rosa Bengala , Tomografía de Coherencia Óptica
13.
Hypertens Res ; 41(1): 8-17, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28978980

RESUMEN

The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. Tubulointerstitial damage with urinary liver-type fatty acid-binding protein (L-FABP) was evaluated using human L-FABP chromosomal transgenic (L-FABP+/-) male mice. The mice in the Ald group (n=7) received systemic Ald infusions via an osmotic minipump and were given 1% NaCl water for 35 days. Those in the Ald-BM group (n=8) were administered BM intraperitoneally in addition to an injection of Ald and salt. The dose of BM was gradually increased every 7 days up to 10 mg kg-1 per day, which was maintained for 14 days. The administration of BM significantly increased renal expression of the Nrf2 target antioxidant gene. Tubulointerstitial damage was significantly ameliorated in the Ald-BM group compared to the Ald group. The increase in reactive oxygen species (ROS) and upregulation of angiotensinogen expression in the kidneys of the Ald group was significantly prevented in the Ald-BM group. The upregulation of human L-FABP expression induced in the kidneys and increase in urinary L-FABP in the Ald group were significantly suppressed by BM administration. In conclusion, BM ameliorated tubulointerstitial damage in the Ald- and salt-induced hypertension model through suppression of both ROS production and intrarenal renin-angiotensin system activation. Urinary L-FABP may be a useful marker reflecting the therapeutic efficacy of BM.


Asunto(s)
Proteínas de Unión a Ácidos Grasos/orina , Hipertensión Renal/prevención & control , Factor 2 Relacionado con NF-E2/metabolismo , Ácido Oleanólico/análogos & derivados , Sustancias Protectoras/uso terapéutico , Sistema Renina-Angiotensina/efectos de los fármacos , Aldosterona/efectos adversos , Angiotensinógeno/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Modelos Animales de Enfermedad , Hipertensión Renal/inducido químicamente , Hipertensión Renal/metabolismo , Riñón/efectos de los fármacos , Riñón/metabolismo , Ratones , Ácido Oleanólico/farmacología , Ácido Oleanólico/uso terapéutico , Estrés Oxidativo/efectos de los fármacos , Sustancias Protectoras/farmacología , Especies Reactivas de Oxígeno/metabolismo , Sistema Renina-Angiotensina/fisiología , Cloruro de Sodio/efectos adversos , Regulación hacia Arriba/efectos de los fármacos
14.
Mol Med Rep ; 16(4): 4994-5000, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28765913

RESUMEN

Heart failure, characterized by impaired systolic and/or diastolic function, is a common cardiovascular disease. The loss of cardiomyocytes due to various factors, including through necrosis or apoptosis can result in heart failure. Previous studies have indicated that excessive aldosterone (ALD) serves an essential role in the process of heart failure, and the heart is also one of the direct targets of ALD, which can provoke hypertrophy and the apoptosis of cardiomyocytes. The aim of the present study was to investigate the protective effect of danshensu (DSS) on ALD­induced cardiomyocytes injury. The present results demonstrated that DSS increased cell viability and decreased the leakage of lactate dehydrogenase in cardiomyocytes exposed to ALD. In addition, DSS decreased the apoptotic rate of ALD­stimulated cells. Further research indicated that DSS­ and cellular tumor antigen p53 (p53)­alone or combination treatment was able to decrease the expression levels of apoptosis regulator BAX and caspase­3, and increase the expression of apoptosis regulator B­cell lymphoma (Bcl)­2 in ALD­stimulated cardiomyocytes. Taken together, the results of the present study suggest that DSS inhibits the harmful effects of ALD on cardiomyocytes via interfering with the p53 signaling pathway. These results provide novel evidence for the potential protective effects of DSS.


Asunto(s)
Aldosterona/efectos adversos , Cardiotónicos/farmacología , Lactatos/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Transducción de Señal/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Apoptosis/efectos de los fármacos , Biomarcadores , Caspasa 3/metabolismo , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , L-Lactato Deshidrogenasa/metabolismo , Masculino , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína X Asociada a bcl-2/metabolismo
15.
Endocrinology ; 158(6): 1859-1874, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28368454

RESUMEN

Primary aldosteronism is characterized by excess aldosterone (ALDO) secretion independent of the renin-angiotensin system and accounts for approximately 10% of hypertension cases. Excess ALDO that is inappropriate for salt intake status causes cardiac hypertrophy, inflammation, fibrosis, and hypertension. The molecular mechanisms that trigger the onset and progression of ALDO-mediated cardiac injury are poorly understood. MicroRNAs (miRNAs) are endogenous, small, noncoding RNAs that have been implicated in diverse cardiac abnormalities, yet very little is known about their regulation and role in ALDO-mediated cardiac injury. To elucidate the regulation of miRNAs in ALDO-mediated cardiac injury, we performed a time-series analysis of left ventricle (LV) miRNA expression. Uninephrectomized male Sprague-Dawley rats were treated with ALDO (0.75 µg/h) infusion and SALT (1.0% NaCl/0.3% KCl) in the drinking water for up to 8 weeks. ALDO/SALT time dependently modulated the expression of multiple miRNAs in the LV. miR-21 was the most upregulated miRNA after 2 weeks of treatment and remained elevated until the end of the study. To elucidate the role of miR-21 in ALDO/SALT-mediated cardiac injury, miR-21 was downregulated by using antagomirs in ALDO/SALT-treated rats. miR-21 downregulation exacerbated ALDO/SALT-mediated cardiac hypertrophy, expression of fibrosis marker genes, interstitial and perivascular fibrosis, OH-proline content, and cardiac dysfunction. These results suggest that ALDO/SALT-mediated cardiac miR-21 upregulation may be a compensatory mechanism that mitigates ALDO/SALT-mediated cardiac deleterious effects. We speculate that miR-21 supplementation would have beneficial effects in reverting or mitigating cardiac injury and dysfunction in patients with primary aldosteronism.


Asunto(s)
Aldosterona/efectos adversos , Cardiopatías/inducido químicamente , Cardiopatías/fisiopatología , Hiperaldosteronismo/complicaciones , MicroARNs/genética , MicroARNs/fisiología , Animales , Regulación de la Expresión Génica , Corazón/efectos de los fármacos , Corazón/fisiopatología , Cardiopatías/genética , Hiperaldosteronismo/metabolismo , Hipertensión/inducido químicamente , Hipertensión/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Cloruro de Sodio/farmacología , Disfunción Ventricular Izquierda/inducido químicamente , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología , Disfunción Ventricular Izquierda/fisiopatología
16.
Curr Hypertens Rep ; 19(5): 40, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28451852

RESUMEN

PURPOSE OF REVIEW: The purpose of this study was to summarize recent findings about cardiovascular benefits and safety of aldosterone blockade in patients with end-stage renal disease (ESRD). RECENT FINDINGS: It is now well recognized that aldosterone's deleterious cardiovascular impact is not limited to its pressor effect arising from an increase in sodium reabsorption in the kidneys. Aldosterone has also been shown to increase blood pressure by a direct activation of the sympathetic nervous system, to cause endothelial and vascular smooth muscle cell dysfunction, myocardial remodeling and fibrosis, and to have pro-arrhythmogenic actions in the heart. These unconventional extra-renal effects of aldosterone make its blockade feasible and potentially beneficial for patients with ESRD. Accumulating data support the idea that aldosterone antagonism leads to a better blood pressure control, reduction in left ventricular (LV) mass, improved LV function, and reduced all-cause and cardiovascular mortality in ESRD patients. Reassuringly, rates of major adverse events, especially, significant hyperkalemia-the most feared adverse consequence-were low with careful patient selection and monitoring.


Asunto(s)
Aldosterona/efectos adversos , Fallo Renal Crónico/fisiopatología , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Aldosterona/fisiología , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Fallo Renal Crónico/tratamiento farmacológico , Receptores de Mineralocorticoides/fisiología
17.
CorSalud ; 8(3)jul.-sept. 2016. graf
Artículo en Español | CUMED | ID: cum-69274

RESUMEN

La aldosterona constituye uno de los componentes del sistema renina-angio-tensina-aldosterona. Existen elementos teóricos y evidencia clínica de su relación con las enfermedades del corazón. Se revisaron numerosas referencias bibliográficas con el objetivo de describir las principales características de la aldosterona y su relación con estas enfermedades. Tradicionalmente se ha pensado que la acción de la aldosterona se restringía tanto a la reabsorción de sodio y agua, como a la excreción de potasio a nivel renal. En años recientes se ha demostrado que desempeña una función primordial en muchos procesos de la patogenia de la enfermedad arterial coronaria y los remodelados cardíaco y vascular(AU)


Asunto(s)
Humanos , Sistema Renina-Angiotensina , Enfermedad de la Arteria Coronaria , Remodelación Ventricular , Aldosterona/efectos adversos , Cardiopatías/etiología
18.
Exp Cell Res ; 346(1): 99-110, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27317889

RESUMEN

Aldosterone has been recognized as a risk factor for the development of chronic kidney disease (CKD). Studies have indicated that enhanced activation of epidermal growth factor receptor (EGFR) is associated with the development and progression of renal fibrosis. But if EGFR is involved in aldosterone-induced renal fibrosis is less investigated. In the present study, we examined the effect of erlotinib, an inhibitor of EGFR tyrosine kinase activity, on the progression of aldosterone-induced renal profibrotic responses in a murine model underwent uninephrectomy. Erlotinib-treated rats exhibited relieved structural lesion comparing with rats treated with aldosterone alone, as characterized by glomerular hypertrophy, mesangial cell proliferation and expansion. Also, erlotinib inhibited the expression of TGF-ß, α-SMA and mesangial matrix proteins such as collagen Ⅳ and fibronectin. In cultured mesangial cells, inhibition of EGFR also abrogated aldosterone-induced expression of extracellular matrix proteins, cell proliferation and migration. We also demonstrated that aldosterone induced the phosphorylation of EGFR through generation of ROS. And the activation of EGFR resulted in the phosphorylation of ERK1/2, leading to the activation of profibrotic pathways. Taken together, we concluded that aldosterone-mediated tissue fibrosis relies on ROS induced EGFR/ERK activation, highlighting EGFR as a potential therapeutic target for modulating renal fibrosis.


Asunto(s)
Aldosterona/efectos adversos , Receptores ErbB/metabolismo , Riñón/metabolismo , Riñón/patología , Transducción de Señal , Animales , Línea Celular , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Receptores ErbB/antagonistas & inhibidores , Clorhidrato de Erlotinib/farmacología , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fibrosis , Hipertrofia , Riñón/efectos de los fármacos , Riñón/enzimología , Glomérulos Renales/efectos de los fármacos , Glomérulos Renales/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/patología , Ratones , Modelos Biológicos , Fosforilación/efectos de los fármacos , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Receptores de Mineralocorticoides/metabolismo , Transducción de Señal/efectos de los fármacos , Activación Transcripcional/genética
19.
Mol Cell Endocrinol ; 429: 84-92, 2016 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-27040306

RESUMEN

Aberrant activation of the renin-angiotensin-aldosterone system (RAAS) contributes to adverse cardiac remodeling and eventual failure. Here we investigated whether TRAF3 Interacting Protein 2 (TRAF3IP2), a redox-sensitive cytoplasmic adaptor molecule and an upstream regulator of nuclear factor-κB (NF-κB) and activator protein-1 (AP-1), mediates aldosterone-induced cardiac hypertrophy and fibrosis. Wild type (WT) and TRAF3IP2-null mice were infused with aldosterone (0.2 mg/kg/day) for 4 weeks along with 1%NaCl in drinking water. Aldosterone/salt, but not salt alone, upregulated TRAF3IP2 expression in WT mouse hearts. Further, aldosterone elevated blood pressure to a similar extent in both WT and TRAF3IP2-null groups. However, TRAF3IP2 gene deletion attenuated aldosterone/salt-induced (i) p65 and c-Jun activation, (ii) extracellular matrix (collagen Iα1 and collagen IIIα1), matrix metalloproteinase (MMP2), lysyl oxidase (LOX), inflammatory cytokine (IL-6 and IL-18), chemokine (CXCL1 and CXCL2), and adhesion molecule (ICAM1) mRNA expression in hearts, (iii) IL-6, IL-18, and MMP2 protein levels, (iv) systemic IL-6 and IL-18 levels, and (iv) cardiac hypertrophy and fibrosis. These results indicate that TRAF3IP2 is a critical signaling intermediate in aldosterone/salt-induced myocardial hypertrophy and fibrosis, and thus a potential therapeutic target in hypertensive heart disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aldosterona/efectos adversos , Cardiomegalia/inducido químicamente , Cardiomegalia/metabolismo , Animales , Presión Sanguínea/efectos de los fármacos , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/genética , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Citocinas/genética , Citocinas/metabolismo , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Mediadores de Inflamación/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Cloruro de Sodio , Factor de Transcripción AP-1/metabolismo
20.
FASEB J ; 29(9): 3899-910, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26054366

RESUMEN

High levels of aldosterone impair renal function by activating proinflammatory and profibrotic pathways. However, the molecular mechanism underlying aldosterone-induced inflammation and fibrosis is unknown. Inflammasome activation contributes to chronic kidney disease. We hypothesized that aldosterone induces renal tubulointerstitial inflammation and fibrosis by activating the inflammasome. Infusing wild-type mice with aldosterone (0.25 mg/kg/d) caused tubulointerstitial damage, increased expression of inflammasome components, caspase 1 activation, and overproduction of IL-1ß and IL-18. These changes were suppressed by eplerenone treatment (100 mg/kg/d) in wild-type mice or in mice deficient in apoptosis-associated speck-like protein with a caspase-recruitment domain (ASC). Caspase 1-positive and F4/80-positive cells colocalized in the interstitium. Bone marrow transplantation using ASC-deficient mice indicated that inflammasome activation in macrophages mediated aldosterone-induced renal fibrosis. IL-18 was detected in culture supernatants of macrophages treated with aldosterone, and mitochondria-derived reactive oxygen species activated the inflammasome in these macrophages. Our results indicate that exposure of macrophages to high levels of aldosterone resulted in the activation of inflammasomes via the mitochondria-derived reactive oxygen species. Thus, inflammasome activation in macrophages may serve as a new therapeutic target for chronic kidney disease.


Asunto(s)
Aldosterona/efectos adversos , Inflamasomas/metabolismo , Enfermedades Renales/metabolismo , Transducción de Señal/efectos de los fármacos , Aldosterona/farmacología , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Adaptadoras de Señalización CARD , Caspasa 1/biosíntesis , Caspasa 1/genética , Activación Enzimática/efectos de los fármacos , Activación Enzimática/genética , Fibrosis , Regulación de la Expresión Génica/efectos de los fármacos , Inflamasomas/genética , Interleucina-18/biosíntesis , Interleucina-18/genética , Interleucina-1beta/biosíntesis , Interleucina-1beta/genética , Enfermedades Renales/inducido químicamente , Enfermedades Renales/genética , Enfermedades Renales/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Noqueados , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...