Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Artículo en Inglés | MEDLINE | ID: mdl-38961842

RESUMEN

Epigenetic mechanisms are considered to contribute to diabetic nephropathy by maintaining memory of poor glycemic control during the early stages of diabetes. However, DNA methylation changes in the human kidney are poorly characterized, because of the lack of cell type-specific analysis. We examined DNA methylation in proximal tubules purified from diabetic nephropathy patients and identified differentially methylated CpG sites, given the critical role of proximal tubules in the kidney injury. Hypermethylation was observed at CpG sites annotated to genes responsible for proximal tubule functions, including gluconeogenesis, nicotinamide adenine dinucleotide synthesis, transporters of glucose, water, phosphate, and drugs, in diabetic kidneys, while genes involved in oxidative stress and the cytoskeleton exhibited demethylation. Methylation levels of CpG sites annotated to ACTN1, BCAR1, MYH9, UBE4B, AFMID, TRAF2, TXNIP, FOXO3, and HNF4A were correlated with the estimated glomerular filtration rate, while methylation of the CpG site in RUNX1 was associated with interstitial fibrosis and tubular atrophy. Hypermethylation of G6PC and HNF4A was accompanied by decreased expression in diabetic kidneys. Proximal tubule-specific hypomethylation of metabolic genes related to HNF4A observed in control kidneys was compromised in diabetic kidneys, suggesting a role for aberrant DNA methylation in the dedifferentiation process. Multiple genes with aberrant DNA methylation in diabetes overlapped genes with altered expressions in maladaptive proximal tubule cells, including transcription factors PPARA and RREB1. In conclusion, DNA methylation derangement in the proximal tubules of patients with diabetes may drive phenotypic changes, characterized by inflammatory and fibrotic features, along with impaired function in metabolism and transport.

2.
J Am Soc Nephrol ; 32(2): 279-289, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33397690

RESUMEN

Hypertension and its comorbidities pose a major public health problem associated with disease-associated factors related to a modern lifestyle, such high salt intake or obesity. Accumulating evidence has demonstrated that aldosterone and its receptor, the mineralocorticoid receptor (MR), have crucial roles in the development of salt-sensitive hypertension and coexisting cardiovascular and renal injuries. Accordingly, clinical trials have repetitively shown the promising effects of MR blockers in these diseases. We and other researchers have identified novel mechanisms of MR activation involved in salt-sensitive hypertension and renal injury, including the obesity-derived overproduction of aldosterone and ligand-independent signaling. Moreover, recent advances in the analysis of cell-specific and context-dependent mechanisms of MR activation in various tissues-including a classic target of aldosterone, aldosterone-sensitive distal nephrons-are now providing new insights. In this review, we summarize recent updates to our understanding of aldosterone-MR signaling, focusing on its role in salt-sensitive hypertension and renal injury.


Asunto(s)
Lesión Renal Aguda/etiología , Aldosterona/fisiología , Hipertensión/etiología , Síndrome Metabólico/etiología , Receptores de Mineralocorticoides/fisiología , Sistema Renina-Angiotensina/fisiología , Humanos , Cloruro de Sodio Dietético
3.
J Am Soc Nephrol ; 31(4): 748-764, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32034107

RESUMEN

BACKGROUND: Regulation of sodium chloride transport in the aldosterone-sensitive distal nephron is essential for fluid homeostasis and BP control. The chloride-bicarbonate exchanger pendrin in ß-intercalated cells, along with sodium chloride cotransporter (NCC) in distal convoluted tubules, complementarily regulate sodium chloride handling, which is controlled by the renin-angiotensin-aldosterone system. METHODS: Using mice with mineralocorticoid receptor deletion in intercalated cells, we examined the mechanism and roles of pendrin upregulation via mineralocorticoid receptor in two different models of renin-angiotensin-aldosterone system activation. We also used aldosterone-treated NCC knockout mice to examine the role of pendrin regulation in salt-sensitive hypertension. RESULTS: Deletion of mineralocorticoid receptor in intercalated cells suppressed the increase in renal pendrin expression induced by either exogenous angiotensin II infusion or endogenous angiotensin II upregulation via salt restriction. When fed a low-salt diet, intercalated cell-specific mineralocorticoid receptor knockout mice with suppression of pendrin upregulation showed BP reduction that was attenuated by compensatory activation of NCC. In contrast, upregulation of pendrin induced by aldosterone excess combined with a high-salt diet was scarcely affected by deletion of mineralocorticoid receptor in intercalated cells, but depended instead on hypokalemic alkalosis through the activated mineralocorticoid receptor-epithelial sodium channel cascade in principal cells. In aldosterone-treated NCC knockout mice showing upregulation of pendrin, potassium supplementation corrected alkalosis and inhibited the pendrin upregulation, thereby lowering BP. CONCLUSIONS: In conjunction with NCC, the two pathways of pendrin upregulation, induced by angiotensin II through mineralocorticoid receptor activation in intercalated cells and by alkalosis through mineralocorticoid receptor activation in principal cells, play important roles in fluid homeostasis during salt depletion and salt-sensitive hypertension mediated by aldosterone excess.


Asunto(s)
Hipertensión/etiología , Nefronas/metabolismo , Nefronas/patología , Receptores de Mineralocorticoides/fisiología , Simportadores del Cloruro de Sodio/fisiología , Transportadores de Sulfato/metabolismo , Aldosterona , Animales , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados , Sistema Renina-Angiotensina/fisiología
4.
Kidney Int ; 98(5): 1242-1252, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32592815

RESUMEN

To investigate the cause of salt sensitivity in a normotensive animal model, we treated rats with a low-dose of the nitric oxide synthase inhibitor, L-NAME, that does not elevate blood pressure per se or induce kidney fibrosis. A high salt diet increased the circulating blood volume both in L-NAME-treated and nontreated animals for the first 24 hours. Thereafter, the blood volume increase persisted only in the L-NAME-treated rats. Blood pressure was higher in the L-NAME-treated group from the start of high salt diet exposure. Within the first 24 hours of salt loading, the L-NAME treated animals failed to show vasodilation and maintained high systemic vascular resistance in response to blood volume expansion. After four weeks on the high salt diet, the slope of the pressure-natriuresis curve was blunted in the L-NAME-treated group. An increase in natriuresis was observed after treatment with hydrochlorothiazide, but not amiloride, a change observed in parallel with increased phosphorylated sodium-chloride cotransporter (NCC). In contrast, a change in blood pressure was not observed in L-NAME-treated NCC-deficient mice fed a high salt diet. Moreover, direct L-NAME-induced NCC activation was demonstrated in cells of the mouse distal convoluted tubule. The vasodilatator, sodium nitroprusside, downregulated phosphorylated NCC expression. The effect of L-NAME on phosphorylated NCC was blocked by both the SPAK inhibitor STOCK2S-26016 and the superoxide dismutase mimetic TEMPO which also attenuated salt-induced hypertension. These results suggest that the initiation of salt sensitivity in normotensive rodents could be due to hyporeactivity of the vasculature and that maintaining blood pressure could result in a high circulating volume due to inappropriate NCC activity in the low-dose L-NAME model. Thus, even slightly impaired nitric oxide production may be important in salt sensitivity regulation in healthy rodents.


Asunto(s)
Hipertensión , Simportadores del Cloruro de Sodio , Animales , Presión Sanguínea , Volumen Sanguíneo , Hipertensión/inducido químicamente , Ratones , NG-Nitroarginina Metil Éster , Ratas , Roedores
5.
Int J Mol Sci ; 21(12)2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32580367

RESUMEN

Renal inflammation is known to be involved in salt-induced renal damage, leading to end-stage renal disease. This study aims to evaluate the role of inflammation in anti-inflammatory and renoprotective effects of beraprost sodium (BPS), a prostaglandin I2 (PGI2) analog, in Dahl salt-sensitive (DS) rats. Five-week-old male DS rats were fed a normal-salt diet (0.5% NaCl), a high-salt diet (8% NaCl), or a high-salt diet plus BPS treatment for 3 weeks. BPS treatment could inhibit marked proteinuria and renal injury in salt-loaded DS rats with elevated blood pressure, accompanied by renal inflammation suppression. Notably, high salt increased renal expression of active Rac1, followed by increased Sgk1 expressions, a downstream molecule of mineralocorticoid receptor (MR) signal, indicating salt-induced activation of Rac1-MR pathway. However, BPS administration inhibited salt-induced Rac1-MR activation as well as renal inflammation and damage, suggesting that Rac1-MR pathway is involved in anti-inflammatory and renoprotective effects of PGI2. Based upon Rac1 activated by inflammation, moreover, BPS inhibited salt-induced activation of Rac1-MR pathway by renal inflammation suppression, resulting in the attenuation of renal damage in salt-loaded DS rats. Thus, BPS is efficacious for the treatment of salt-induced renal injury.


Asunto(s)
Lesión Renal Aguda/prevención & control , Epoprostenol/análogos & derivados , Regulación de la Expresión Génica/efectos de los fármacos , Inflamación/prevención & control , Receptores de Mineralocorticoides/metabolismo , Cloruro de Sodio/toxicidad , Proteína de Unión al GTP rac1/metabolismo , Lesión Renal Aguda/inducido químicamente , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Animales , Epoprostenol/farmacología , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratas , Ratas Sprague-Dawley , Receptores de Mineralocorticoides/genética , Vasodilatadores/farmacología , Proteína de Unión al GTP rac1/genética
6.
J Am Soc Nephrol ; 29(1): 57-68, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29021385

RESUMEN

The renin-angiotensin-aldosterone system has an important role in the control of fluid homeostasis and BP during volume depletion. Dietary salt restriction elevates circulating angiotensin II (AngII) and aldosterone levels, increasing levels of the Cl-/HCO3- exchanger pendrin in ß-intercalated cells and the Na+-Cl- cotransporter (NCC) in distal convoluted tubules. However, the independent roles of AngII and aldosterone in regulating these levels remain unclear. In C57BL/6J mice receiving a low-salt diet or AngII infusion, we evaluated the membrane protein abundance of pendrin and NCC; assessed the phosphorylation of the mineralocorticoid receptor, which selectively inhibits aldosterone binding in intercalated cells; and measured BP by radiotelemetry in pendrin-knockout and wild-type mice. A low-salt diet or AngII infusion upregulated NCC and pendrin levels, decreased the phosphorylation of mineralocorticoid receptor in ß-intercalated cells, and increased plasma aldosterone levels. Notably, a low-salt diet did not alter BP in wild-type mice, but significantly decreased BP in pendrin-knockout mice. To dissect the roles of AngII and aldosterone, we performed adrenalectomies in mice to remove aldosterone from the circulation. In adrenalectomized mice, AngII infusion again upregulated NCC expression, but did not affect pendrin expression despite the decreased phosphorylation of mineralocorticoid receptor. By contrast, AngII and aldosterone coadministration markedly elevated pendrin levels in adrenalectomized mice. Our results indicate that aldosterone is necessary for AngII-induced pendrin upregulation, and suggest that pendrin contributes to the maintenance of normal BP in cooperation with NCC during activation of the renin-angiotensin-aldosterone system by dietary salt restriction.


Asunto(s)
Aldosterona/sangre , Angiotensina II/farmacología , Simportadores del Cloruro de Sodio/metabolismo , Transportadores de Sulfato/metabolismo , Vasoconstrictores/farmacología , Adrenalectomía , Aldosterona/farmacología , Animales , Presión Sanguínea/genética , Túbulos Renales Distales/citología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Receptores de Mineralocorticoides/metabolismo , Cloruro de Sodio Dietético/administración & dosificación , Transportadores de Sulfato/genética , Regulación hacia Arriba/efectos de los fármacos
7.
Am J Physiol Renal Physiol ; 314(4): F551-F560, 2018 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212764

RESUMEN

Epigenetic abnormalities have been suggested to mediate metabolic memory observed in diabetic complications. We have shown that epigenetic alterations may induce persistent phenotypic changes in the proximal tubules of the diabetic kidneys. In this study, we show that pregnane X receptor (PXR), a xenobiotic nuclear receptor, is epigenetically altered and upregulated and may have a possible function in the diabetic kidney. PXR has been shown to play a critical role in metabolic changes in obesity and diabetes; however, its distribution and function in the kidney are unknown. In the normal kidney, Pxr was selectively expressed in the proximal tubular cells with demethylation in the promoter DNA. In db/db mice, significant increases in Pxr mRNA, further demethylation of DNA, and stimulatory histone marks in the promoter were observed. Epigenetic changes are likely to play a causative role in PXR induction, since a DNA methyltransferase inhibitor increased PXR mRNA in cultured human proximal tubular cells. Administration of a PXR agonist increased mRNA levels of solute carrier organic anion transporter family member 2B1 ( Slco2b1), a xenobiotic transporter; response gene to complement 32 ( Rgc32), a molecule known to exert fibrotic effects in the kidney; and phosphoenolpyruvate carboxykinase 1 ( Pck1), a gluconeogenic enzyme in the kidney. The expressions of these genes were inhibited by PXR small interfering RNA in cultured proximal tubular cells. Increased mRNA levels of Slco2b1, Rgc32, and Pck1 were also observed in the kidney of db/db mice. These data indicate that PXR is upregulated in the diabetic kidney with aberrant epigenetic modifications and may modulate the course of diabetic kidney disease through the activation of these genes.


Asunto(s)
Metilación de ADN , Nefropatías Diabéticas/genética , Metabolismo Energético/genética , Epigénesis Genética , Túbulos Renales Proximales/metabolismo , Receptor X de Pregnano/genética , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Nefropatías Diabéticas/metabolismo , Modelos Animales de Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Fenotipo , Fosfoenolpiruvato Carboxiquinasa (GTP)/genética , Fosfoenolpiruvato Carboxiquinasa (GTP)/metabolismo , Receptor X de Pregnano/metabolismo , Regiones Promotoras Genéticas
8.
J Am Soc Nephrol ; 26(10): 2388-97, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25653098

RESUMEN

Epigenetic mechanisms may underlie the progression of diabetic kidney disease. Because the kidney is a heterogeneous organ with different cell types, we investigated DNA methylation status of the kidney in a cell type-specific manner. We first identified genes specifically demethylated in the normal proximal tubules obtained from control db/m mice, and next delineated the candidate disease-modifying genes bearing aberrant DNA methylation induced by diabetes using db/db mice. Genes involved in glucose metabolism, including Sglt2, Pck1, and G6pc, were selectively hypomethylated in the proximal tubules in control mice. Hnf4a, a transcription factor regulating transporters for reabsorption, was also selectively demethylated. In diabetic mice, aberrant hypomethylation of Agt, Abcc4, Cyp4a10, Glut5, and Met and hypermethylation of Kif20b, Cldn18, and Slco1a1 were observed. Time-dependent demethylation of Agt, a marker of diabetic kidney disease, was accompanied by histone modification changes. Furthermore, inhibition of DNA methyltransferase or histone deacetylase increased Agt mRNA in cultured human proximal tubular cells. Aberrant DNA methylation and concomitant changes in histone modifications and mRNA expression in the diabetic kidney were resistant to antidiabetic treatment with pioglitazone. These results suggest that an epigenetic switch involving aberrant DNA methylation causes persistent mRNA expression of select genes that may lead to phenotype changes of the proximal tubules in diabetic kidney disease.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Túbulos Renales Proximales/metabolismo , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
9.
Curr Hypertens Rep ; 17(6): 552, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25903070

RESUMEN

The impaired capacity of the kidney to excrete sodium plays an essential role in the development of hypertension. Adrenal corticosteroids control renal handling of sodium by regulating tubular sodium reabsorption in the distal nephron where both mineralocorticoid receptors (MR) and glucocorticoid receptors are expressed. In addition, cell type- and segment-specific expression of 11ß-HSD2 and sodium transporters such as Na-Cl cotransporter (NCC), epithelial sodium channel (ENaC), and pendrin/Na(+)-driven Cl(-)/HCO3 (-) exchanger (NDCBE) builds a distinctive model of sodium transport in the aldosterone-sensitive distal nephron. Aberrant MR activation in the distal nephron triggers salt-sensitive hypertension and hypokalemia through inappropriate sodium reabsorption and potassium secretion. However, MR activity is not necessarily modulated by the ligand alone. Recently, several lines of evidence revealed alternative mechanisms that regulate the activity of MR in a ligand-independent manner or through ligand binding modulation. This review summarizes the disorders related to MR activation in individual tubular cells and highlights the renal mechanism of salt-sensitive hypertension and new approaches for the prevention and treatment of this disease.


Asunto(s)
Hipertensión/metabolismo , Receptores de Mineralocorticoides/metabolismo , Cloruro de Sodio Dietético/metabolismo , Animales , Humanos , Riñón/lesiones , Riñón/metabolismo , Mineralocorticoides/metabolismo
10.
Nephron Exp Nephrol ; 126(1): 16-24, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24603367

RESUMEN

BACKGROUND/AIMS: Obesity and diabetes are intimately interrelated, and are independent risk factors for kidney disease. Overactivation of mineralocorticoid receptor (MR) is implicated in end organ damage of both pathologies. But the underlying mechanism of MR activation in kidney remains uncertain. We explored the involvement of Rac1, which we previously identified as a ligand-independent MR activator, in renal MR activation in vitro and in vivo. METHODS: We evaluated the MR activity and Rac1 activity under high-glucose stimulation using luciferase reporter system and glutathione S-transferase pull-down assay in cultured mesangial cells. To elucidate the role of Rac1 in vivo, we employed KKA(y), a mouse model of obesity-related type 2 diabetes, which spontaneously developed massive albuminuria and distinct glomerular lesions accompanied by increased plasma aldosterone concentration. RESULTS: High-glucose stimulation increased Rac1 activity and MR transcriptional activity in cultured mesangial cells. Overexpression of constitutively active Rac1 activated MR, and glucose-induced MR activation was suppressed by overexpression of dominant negative Rac1 or Rac inhibitor EHT1864. In KKA(y), renal Rac1 was activated, and nuclear MR was increased. EHT1864 treatment suppressed renal Rac1 and MR activity and mitigated renal pathology of KKA(y) without changing plasma aldosterone concentration. CONCLUSION: Our results suggest that MR activation plays an important role in the nephropathy of KKA(y) mice, and that glucose-induced Rac1 activation, in addition to hyperaldosteronemia, contributes to their renal MR activation. Along with MR blockade, Rac inhibition may potentially be a preferred option in the treatment of nephropathy in obesity-related diabetic patients.


Asunto(s)
Nefropatías Diabéticas/metabolismo , Obesidad/metabolismo , Receptores de Mineralocorticoides/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Albuminuria/metabolismo , Albuminuria/prevención & control , Animales , Western Blotting , Línea Celular , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Nefropatías Diabéticas/etiología , Nefropatías Diabéticas/genética , Glucosa/farmacología , Humanos , Inmunohistoquímica , Riñón/efectos de los fármacos , Riñón/metabolismo , Riñón/patología , Masculino , Células Mesangiales/efectos de los fármacos , Células Mesangiales/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/complicaciones , Obesidad/genética , Tamaño de los Órganos/efectos de los fármacos , Pironas/farmacología , Quinolinas/farmacología , Receptores de Mineralocorticoides/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Proteína de Unión al GTP rac1/genética
11.
Clin Exp Nephrol ; 18(4): 593-9, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24154707

RESUMEN

BACKGROUND: The renin-angiotensin-aldosterone system (RAAS) plays pivotal roles in the pathogenesis of chronic kidney disease (CKD) progression. Aliskiren, a direct renin inhibitor, inhibits the rate-limiting step of the RAAS without any alternative pathway. It is proven to reduce albuminuria in CKD patients treated with angiotensin blockade. However, there are few reports which evaluate the advantage of aliskiren as the first-line drug against CKD progression in RAAS-activated hypertensive patients. METHODS: Tsukuba hypertensive mice (THM), double transgenic mice carrying both the human renin and human angiotensinogen genes, were fed a high-salt diet and treated with hydraladine, ramipril and aliskiren for 10 weeks. Blood pressure and urinary albumin excretion were measured every 2 weeks during the experimental period. We evaluated renal histological changes and gene expression. Plasma angiotensin concentration was measured to evaluate the RAAS inhibitory effect. RESULTS: High-salt-loaded THM showed severe hypertension and renal injury. All antihypertensive drugs suppressed blood pressure and prevented renal disease progression. RAAS blockade showed a higher renoprotective effect than hydraladine despite an equivalent blood pressure lowering effect. Aliskiren exhibited even stronger renoprotection than ramipril. Plasma angiotensin concentration was increased in THM fed both normal salt and high salt. Hydraladine did not alter the plasma angiotensin concentration. Ramipril significantly decreased angiotensin II concentration. Aliskiren treatment almost completely suppressed angiotensin I and resulted in lower angiotensin II concentration than ramipril treatment. CONCLUSION: Aliskiren prevents renal disease progression by suppressing both angiotensin I and II in RAAS-activated pathology. Our data suggest the application of a renin inhibitor for preventing kidney disease progression in CKD patients.


Asunto(s)
Amidas/farmacología , Angiotensina II/sangre , Angiotensina I/sangre , Angiotensinógeno/metabolismo , Antihipertensivos/farmacología , Fumaratos/farmacología , Hipertensión/tratamiento farmacológico , Enfermedades Renales/prevención & control , Riñón/efectos de los fármacos , Sistema Renina-Angiotensina/efectos de los fármacos , Renina/antagonistas & inhibidores , Cloruro de Sodio Dietético , Albuminuria/tratamiento farmacológico , Albuminuria/metabolismo , Inhibidores de la Enzima Convertidora de Angiotensina/farmacología , Angiotensinógeno/genética , Animales , Presión Sanguínea/efectos de los fármacos , Citoprotección , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Regulación hacia Abajo , Humanos , Hidralazina/farmacología , Hipertensión/sangre , Hipertensión/genética , Hipertensión/patología , Hipertensión/fisiopatología , Riñón/metabolismo , Riñón/patología , Riñón/fisiopatología , Enfermedades Renales/sangre , Enfermedades Renales/genética , Enfermedades Renales/patología , Enfermedades Renales/fisiopatología , Ratones , Ratones Transgénicos , Ramipril/farmacología , Renina/genética , Renina/metabolismo , Sistema Renina-Angiotensina/genética , Factores de Tiempo
12.
J Am Soc Nephrol ; 23(6): 997-1007, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22440899

RESUMEN

Experiments with hyperaldosteronemic animals suggest that, despite lowering plasma aldosterone, salt worsens renal injury by paradoxical activation of the mineralocorticoid receptor (MR). Salt and aldosterone synergistically contribute to renal impairment through Rac1-mediated activation of the MR, but whether angiotensin II also promotes renal injury through this mechanism is unknown. Here, we placed angiotensin II-overproducing double transgenic Tsukuba hypertensive mice on a low- or high-salt intake for 6 weeks and treated some animals with adrenalectomy, the MR antagonist eplerenone, the Rac inhibitor EHT1864, or hydralazine. High-salt intake, but not low-salt intake, led to hypertension and prominent kidney injury. Adrenalectomy prevented angiotensin II/salt-induced nephropathy in mice receiving high-salt intake, which was recapitulated by aldosterone supplementation, suggesting the involvement of aldosterone/MR signaling. Plasma aldosterone levels, however, were lower in high- than low-salt conditions. Instead, angiotensin II/salt-evoked MR activation associated with Rac1 activation and was not dependent on plasma aldosterone level. Both EHT1864 and eplerenone repressed the augmented MR signaling and mitigated kidney injury with partial but significant reduction in BP with high-salt intake. Hydralazine similarly reduced BP, but it neither suppressed the Rac1-MR pathway nor ameliorated the nephropathy. Taken together, these results show that angiotensin II and salt accelerate kidney injury through Rac1-mediated MR activation. Rac inhibition may be a promising strategy for the treatment of CKD.


Asunto(s)
Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Aldosterona/metabolismo , Angiotensina II/efectos adversos , Receptores de Mineralocorticoides/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Lesión Renal Aguda/fisiopatología , Adrenalectomía/métodos , Aldosterona/sangre , Análisis de Varianza , Angiotensina II/farmacología , Animales , Western Blotting , Modelos Animales de Enfermedad , Eplerenona , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Transducción de Señal , Cloruro de Sodio Dietético/farmacología , Espironolactona/análogos & derivados , Espironolactona/farmacología , Estadísticas no Paramétricas , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
13.
Sci Rep ; 12(1): 20310, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36434091

RESUMEN

Development of the renal medulla continues after birth to form mature renal papilla and obtain urine-concentrating ability. Here, we found that a small GTPase, Rac1, plays a critical role in the postnatal development of renal papilla. Mice with distal tubule-specific deletion of Rac1 reached adulthood but showed polydipsia and polyuria with an impaired ability to concentrate urine. The elongation of renal papilla that occurs in the first weeks after birth was impaired in the Rac1-deficient infants, resulting in shortening and damage of the renal papilla. Moreover, the osmoprotective signaling mediated by nuclear factor of activated T cells 5, which is a key molecule of osmotic response to osmotic stress in renal medulla, was significantly impaired in the kidneys of the Rac1-deficient infants. These results demonstrate that Rac1 plays an important role in the development of renal papilla in the postnatal period, and suggested a potential link between Rac1 and osmotic response.


Asunto(s)
Médula Renal , Riñón , Ratones , Animales , Transducción de Señal
14.
Hypertension ; 78(1): 82-93, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34058848
15.
J Clin Invest ; 130(8): 4152-4166, 2020 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-32597829

RESUMEN

Aging is associated with a high prevalence of hypertension due to elevated susceptibility of BP to dietary salt, but its mechanism is unknown. Serum levels of Klotho, an anti-aging factor, decline with age. We found that high salt (HS) increased BP in aged mice and young heterozygous Klotho-knockout mice and was associated with increased vascular expression of Wnt5a and p-MYPT1, which indicate RhoA activity. Not only the Wnt inhibitor LGK974 and the Wnt5a antagonist Box5 but Klotho supplementation inhibits HS-induced BP elevation, similarly to the Rho kinase inhibitor fasudil, associated with reduced p-MYPT1 expression in both groups of mice. In cultured vascular smooth muscle cells, Wnt5a and angiotensin II (Ang II) increased p-MYPT1 expression but knockdown of Wnt5a with siRNA abolished Ang II-induced upregulation of p-MYPT1, indicating that Wnt5a is indispensable for Ang II-induced Rho/ROCK activation. Notably, Klotho inhibited Wnt5a- and Ang II-induced upregulation of p-MYPT1. Consistently, Klotho supplementation ameliorated HS-induced augmentation of reduced renal blood flow (RBF) response to intra-arterial infusion of Ang II and the thromboxane A2 analog U46619, which activated RhoA in both groups of mice and were associated with the inhibition of BP elevation, suggesting that abnormal response of RBF to Ang II contributes to HS-induced BP elevation. Thus, Klotho deficiency underlies aging-associated salt-sensitive hypertension through vascular non-canonical Wnt5a/RhoA activation.


Asunto(s)
Envejecimiento , Glucuronidasa/deficiencia , Hipertensión , Músculo Liso Vascular , Miocitos del Músculo Liso , Cloruro de Sodio Dietético/efectos adversos , Proteína Wnt-5a/metabolismo , Envejecimiento/efectos de los fármacos , Envejecimiento/genética , Envejecimiento/metabolismo , Envejecimiento/patología , Angiotensina II/genética , Angiotensina II/metabolismo , Animales , Glucuronidasa/metabolismo , Hipertensión/inducido químicamente , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/patología , Proteínas Klotho , Masculino , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Fosfatasa de Miosina de Cadena Ligera/genética , Fosfatasa de Miosina de Cadena Ligera/metabolismo , Cloruro de Sodio Dietético/farmacología , Proteína Wnt-5a/genética
16.
Artículo en Inglés | MEDLINE | ID: mdl-32883689

RESUMEN

INTRODUCTION: Renal tubular injury contributes to the decline in kidney function in patients with diabetes. Cell type-specific DNA methylation patterns have been used to calculate proportions of particular cell types. In this study, we developed a method to detect renal tubular injury in patients with diabetes by detecting exfoliated tubular cells shed into the urine based on tubular cell-specific DNA methylation patterns. RESEARCH DESIGN AND METHODS: We identified DNA methylation patterns specific for human renal proximal tubular cells through compartment-specific methylome analysis. We next determined the methylation levels of proximal tubule-specific loci in urine sediment of patients with diabetes and analyzed correlation with clinical variables. RESULTS: We identified genomic loci in SMTNL2 and G6PC to be selectively unmethylated in human proximal tubular cells. The methylation levels of SMTNL2 and G6PC in urine sediment, deemed to reflect the proportion of exfoliated proximal tubular cells due to injury, correlated well with each other. Methylation levels of SMTNL2 in urine sediment significantly correlated with the annual decline in estimated glomerular filtration rate. Moreover, addition of urinary SMTNL2 methylation to a model containing known risk factors significantly improved discrimination of patients with diabetes with faster estimated glomerular filtration rate decline. CONCLUSIONS: This study demonstrates that patients with diabetes with continual loss in kidney function may be stratified by a specific DNA methylation signature through epigenetic urinalysis and provides further evidence at the level of exfoliated cells in the urine that injury of proximal tubular cells may contribute to pathogenesis of diabetic kidney disease.


Asunto(s)
Diabetes Mellitus , Nefropatías Diabéticas , ADN/metabolismo , Diabetes Mellitus/genética , Diabetes Mellitus/metabolismo , Nefropatías Diabéticas/diagnóstico , Nefropatías Diabéticas/genética , Tasa de Filtración Glomerular , Humanos , Riñón/metabolismo , Metilación
17.
Hypertens Res ; 42(4): 514-521, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30631161

RESUMEN

Excessive dietary salt intake can counteract the renoprotective effects of renin-angiotensin system (RAS) blockade in hypertensive patients with chronic kidney disease (CKD). In rodents, salt loading induces hypertension and renal damage by activating the mineralocorticoid receptor (MR) independently of plasma aldosterone levels. Thus, high salt-induced resistance to RAS blockade may be mediated by MR activation. To test this, a post hoc analysis of the Eplerenone Combination Versus Conventional Agents to Lower Blood Pressure on Urinary Antialbuminuric Treatment Effect (EVALUATE) trial was conducted. Thus, 304 non-diabetic hypertensive patients on RAS-blocking therapy were divided into tertiles according to salt intake (estimated 24-h urinary sodium excretion at baseline) and compared in terms of percent reduction in urinary albumin-to-creatinine ratio (UACR) at 52 weeks relative to baseline. The eplerenone-treated patients in the highest sodium excretion tertile exhibited significantly greater reduction in UACR than the placebo subjects in the same tertile (-22.5% vs. +21.8%, p = 0.02). This disparity was not observed in the lowest (-10.2% vs. -0.84%, p = 0.65) or middle (-19.5% vs. +9.5%, p = 0.22) tertiles. Similar systolic blood pressure changes were observed. In the whole cohort, reduction in UACR correlated positively with reduction in systolic blood pressure (r2 = 0.04, p = 0.02). These results support the hypothesis that excessive salt intake can enhance resistance to RAS blockade by activating MR. They also suggest that eplerenone plus RAS blockade may be effective for CKD in hypertensive patients, especially those with excessive salt intake.


Asunto(s)
Albuminuria/tratamiento farmacológico , Presión Sanguínea/efectos de los fármacos , Eplerenona/uso terapéutico , Hipertensión/complicaciones , Antagonistas de Receptores de Mineralocorticoides/uso terapéutico , Adulto , Anciano , Albuminuria/complicaciones , Albuminuria/fisiopatología , Presión Sanguínea/fisiología , Eplerenona/farmacología , Femenino , Humanos , Hipertensión/fisiopatología , Masculino , Persona de Mediana Edad , Antagonistas de Receptores de Mineralocorticoides/farmacología , Sistema Renina-Angiotensina/efectos de los fármacos , Cloruro de Sodio Dietético , Adulto Joven
18.
JCI Insight ; 3(21)2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30385711

RESUMEN

Maternal malnutrition, which causes prenatal exposure to excessive glucocorticoid, induces adverse metabolic programming, leading to hypertension in offspring. In offspring of pregnant rats receiving a low-protein diet or dexamethasone, a synthetic glucocorticoid, mRNA expression of angiotensin receptor type 1a (Agtr1a) in the paraventricular nucleus (PVN) of the hypothalamus was upregulated, concurrent with reduced expression of DNA methyltransferase 3a (Dnmt3a), reduced binding of DNMT3a to the Agtr1a gene, and DNA demethylation. Salt loading increased BP in both types of offspring, suggesting that elevated hypothalamic Agtr1a expression is epigenetically modulated by excessive glucocorticoid and leads to adult-onset salt-sensitive hypertension. Consistent with this, dexamethasone treatment of PVN cells upregulated Agtr1a, while downregulating Dnmt3a, and decreased DNMT3a binding and DNA demethylation at the Agtr1a locus. In addition, Dnmt3a knockdown upregulated Agtr1a independently of dexamethasone. Hypothalamic neuron-specific Dnmt3a-deficient mice exhibited upregulation of Agtr1a in the PVN and salt-induced BP elevation without dexamethasone treatment. By contrast, dexamethasone-treated Agtr1a-deficient mice failed to show salt-induced BP elevation, despite reduced expression of Dnmt3a. Thus, epigenetic modulation of hypothalamic angiotensin signaling contributes to salt-sensitive hypertension induced by prenatal glucocorticoid excess in offspring of mothers that are malnourished during pregnancy.


Asunto(s)
Metilación de ADN/genética , Hipertensión/genética , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Angiotensina Tipo 1/metabolismo , Animales , Animales Recién Nacidos , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A , Dexametasona/provisión & distribución , Epigenómica , Femenino , Glucocorticoides/provisión & distribución , Hipertensión/metabolismo , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal , Desnutrición Proteico-Calórica/complicaciones , ARN Mensajero/genética , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/fisiología
19.
Sci Rep ; 8(1): 16338, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30397232

RESUMEN

Epigenetic modulation may underlie the progression of diabetic nephropathy (DN). Involvement of TGFB1 in mesangial fibrosis of DN led us to hypothesize that Tgfb1 DNA demethylation contributes to progression of DN. In primary mesangial cells from diabetic (db/db) mouse kidneys, demethylation of Tgfb1 DNA and upregulation of Tgfb1 mRNA progressed simultaneously. USF1 binding site in Tgfb1 promoter region were demethylated, and binding of USF1 increased, with decreased binding of DNMT1 in db/db compared with control. Given downregulation of Tgfb1 expression by folic acid, antioxidant Tempol reversed DNA demethylation, with increased and decreased recruitment of DNMT1 and USF1 to the promoter, resulting in decreased Tgfb1 expression in db/db mice. Addition of H2O2 to mesangial cells induced DNA demethylation and upregulated Tgfb1 expression. Finally, Tempol attenuated mesangial fibrosis in db/db mice. We conclude that aberrant DNA methylation of Tgfb1 due to ROS overproduction play a key to mesangial fibrosis during DN progression.


Asunto(s)
Metilación de ADN , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Células Mesangiales/metabolismo , Factor de Crecimiento Transformador beta1/genética , Animales , Diabetes Mellitus Experimental/metabolismo , Progresión de la Enfermedad , Fibrosis , Masculino , Células Mesangiales/patología , Ratones , Ratones Endogámicos C57BL , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Regulación hacia Arriba/genética
20.
Hypertension ; 70(1): 111-118, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28559392

RESUMEN

Genome-wide analysis of renal sodium-transporting system has identified specific variations of Mendelian hypertensive disorders, including HSD11B2 gene variants in apparent mineralocorticoid excess. However, these genetic variations in extrarenal tissue can be involved in developing hypertension, as demonstrated in former studies using global and brain-specific Hsd11b2 knockout rodents. To re-examine the importance of renal dysfunction on developing hypertension, we generated kidney-specific Hsd11b2 knockout mice. The knockout mice exhibited systemic hypertension, which was abolished by reducing salt intake, suggesting its salt-dependency. In addition, we detected an increase in renal membrane expressions of cleaved epithelial sodium channel-α and T53-phosphorylated Na+-Cl- cotransporter in the knockout mice. Acute intraperitoneal administration of amiloride-induced natriuresis and increased urinary sodium/potassium ratio more in the knockout mice compared with those in the wild-type control mice. Chronic administration of amiloride and high-KCl diet significantly decreased mean blood pressure in the knockout mice, which was accompanied with the correction of hypokalemia and the resultant decrease in Na+-Cl- cotransporter phosphorylation. Accordingly, a Na+-Cl- cotransporter blocker hydrochlorothiazide significantly decreased mean blood pressure in the knockout mice. Chronic administration of mineralocorticoid receptor antagonist spironolactone significantly decreased mean blood pressure of the knockout mice along with downregulation of cleaved epithelial sodium channel-α and phosphorylated Na+-Cl- cotransporter expression in the knockout kidney. Our data suggest that kidney-specific deficiency of 11ß-HSD2 leads to salt-dependent hypertension, which is attributed to mineralocorticoid receptor-epithelial sodium channel-Na+-Cl- cotransporter activation in the kidney, and provides evidence that renal dysfunction is essential for developing the phenotype of apparent mineralocorticoid excess.


Asunto(s)
11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/genética , Presión Sanguínea/genética , Hipertensión , Receptores de Mineralocorticoides/metabolismo , Insuficiencia Renal , Cloruro de Sodio Dietético , Animales , Modelos Animales de Enfermedad , Canales Epiteliales de Sodio/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Eliminación de Gen , Hipertensión/genética , Hipertensión/metabolismo , Hipertensión/fisiopatología , Hipertensión/terapia , Transporte Iónico/efectos de los fármacos , Ratones , Ratones Noqueados , Antagonistas de Receptores de Mineralocorticoides/farmacología , Insuficiencia Renal/metabolismo , Insuficiencia Renal/fisiopatología , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio Dietético/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA