Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Physiol Rep ; 9(20): e15080, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34665521

RESUMEN

The nitric oxide (NO)-generating enzyme, NO synthase-1ß (NOS1ß), is essential for sodium (Na+ ) homeostasis and blood pressure control. We previously showed that collecting duct principal cell NOS1ß is critical for inhibition of the epithelial sodium channel (ENaC) during high Na+ intake. Previous studies on freshly isolated cortical collecting ducts (CCD) demonstrated that exogenous NO promotes basolateral potassium (K+ ) conductance through basolateral channels, presumably Kir 4.1 (Kcnj10) and Kir 5.1 (Kcnj16). We, therefore, investigated the effects of NOS1ß knockout on Kir 4.1/Kir 5.1 channel activity. Indeed, in CHO cells overexpressing NOS1ß and Kir 4.1/Kir 5.1, the inhibition of NO signaling decreased channel activity. Male littermate control and principal cell NOS1ß knockout mice (CDNOS1KO) on a 7-day, 4% NaCl diet (HSD) were used to detect changes in basolateral K+ conductance. We previously demonstrated that CDNOS1KO mice have high circulating aldosterone despite a high-salt diet and appropriately suppressed renin. We observed greater Kir 4.1 cortical abundance and significantly greater Kir 4.1/Kir 5.1 single-channel activity in the principal cells from CDNOS1KO mice. Moreover, blocking aldosterone action with in vivo spironolactone treatment resulted in lower Kir 4.1 abundance and greater plasma K+ in the CDNOS1KO mice compared to controls. Lowering K+ content in the HSD prevented the high aldosterone and greater plasma Na+ of CDNOS1KO mice and normalized Kir 4.1 abundance. We conclude that during chronic HSD, lack of NOS1ß leads to increased plasma K+ , enhanced circulating aldosterone, and activation of ENaC and Kir 4.1/Kir 5.1 channels. Thus, principal cell NOS1ß is required for the regulation of both Na+ and K+ by the kidney.


Asunto(s)
Homeostasis , Túbulos Renales Colectores/metabolismo , Óxido Nítrico Sintasa de Tipo I/fisiología , Canales de Potasio de Rectificación Interna/metabolismo , Potasio/metabolismo , Sodio/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Transporte Iónico , Masculino , Ratones , Ratones Noqueados , Canales de Potasio de Rectificación Interna/genética
2.
JCI Insight ; 5(16)2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32673289

RESUMEN

Histone deacetylase (HDAC) enzymes regulate transcription through epigenetic modification of chromatin structure, but their specific functions in the kidney remain elusive. We discovered that the human kidney expresses class I HDACs. Kidney medulla-specific inhibition of class I HDACs in the rat during high-salt feeding results in hypertension, polyuria, hypokalemia, and nitric oxide deficiency. Three new inducible murine models were used to determine that HDAC1 and HDAC2 in the kidney epithelium are necessary for maintaining epithelial integrity and maintaining fluid-electrolyte balance during increased dietary sodium intake. Moreover, single-nucleus RNA-sequencing determined that epithelial HDAC1 and HDAC2 are necessary for expression of many sodium or water transporters and channels. In performing a systematic review and meta-analysis of serious adverse events associated with clinical HDAC inhibitor use, we found that HDAC inhibitors increased the odds ratio of experiencing fluid-electrolyte disorders, such as hypokalemia. This study provides insight on the mechanisms of potential serious adverse events with HDAC inhibitors, which may be fatal to critically ill patients. In conclusion, kidney tubular HDACs provide a link between the environment, such as consumption of high-salt diets, and regulation of homeostatic mechanisms to remain in fluid-electrolyte balance.


Asunto(s)
Electrólitos/metabolismo , Inhibidores de Histona Desacetilasas/efectos adversos , Histona Desacetilasas/metabolismo , Médula Renal/metabolismo , Animales , Benzamidas/farmacología , Presión Sanguínea/efectos de los fármacos , Femenino , Histona Desacetilasa 1/genética , Histona Desacetilasa 1/metabolismo , Histona Desacetilasa 2/genética , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/genética , Homeostasis/efectos de los fármacos , Homeostasis/fisiología , Humanos , Médula Renal/efectos de los fármacos , Médula Renal/fisiopatología , Masculino , Óxido Nítrico/metabolismo , Potasio/sangre , Piridinas/farmacología , Ratas Sprague-Dawley , Cloruro de Sodio Dietético/farmacología , Equilibrio Hidroelectrolítico/fisiología
3.
Am J Physiol Renal Physiol ; 317(3): F547-F559, 2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31241990

RESUMEN

The collecting duct (CD) concentrates the urine, thereby maintaining body water volume and plasma osmolality within a normal range. The endocrine hormone arginine vasopressin acts in the CD to increase water permeability via the vasopressin 2 receptor (V2R)-aquaporin (AQP) axis. Recent studies have suggested that autocrine factors may also contribute to the regulation of CD water permeability. Nitric oxide is produced predominantly by nitric oxide synthase 1 (NOS1) in the CD and acts as a diuretic during salt loading. The present study sought to determine whether CD NOS1 regulates diuresis during changes in hydration status. Male and female control and CD NOS1 knockout (CDNOS1KO) mice were hydrated (5% sucrose water), water deprived, or acutely challenged with the V2R agonist desmopressin. In male mice, water deprivation resulted in decreased urine flow and increased plasma osmolality, copeptin concentration, and kidney AQP2 abundance independent of CD NOS1. In female control mice, water deprivation reduced urine flow, increased plasma osmolality and copeptin, but did not significantly change total AQP2; however, there was increased basolateral AQP3 localization. Surprisingly, female CDNOS1KO mice while on the sucrose water presented with symptoms of dehydration. Fibroblast growth factor 21, an endocrine regulator of sweetness preference, was significantly higher in female CDNOS1KO mice, suggesting that this was reducing their drive to drink the sucrose water. With acute desmopressin challenge, female CDNOS1KO mice failed to appropriately concentrate their urine, resulting in higher plasma osmolality than controls. In conclusion, CD NOS1 plays only a minor role in urine-concentrating mechanisms.


Asunto(s)
Deshidratación/enzimología , Diuresis , Capacidad de Concentración Renal , Túbulos Renales Colectores/enzimología , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico/metabolismo , Animales , Fármacos Antidiuréticos/farmacología , Acuaporina 2/genética , Acuaporina 2/metabolismo , Acuaporina 3/genética , Acuaporina 3/metabolismo , Desamino Arginina Vasopresina/farmacología , Deshidratación/fisiopatología , Modelos Animales de Enfermedad , Diuresis/efectos de los fármacos , Femenino , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Capacidad de Concentración Renal/efectos de los fármacos , Túbulos Renales Colectores/efectos de los fármacos , Masculino , Ratones Noqueados , Óxido Nítrico Sintasa de Tipo I/deficiencia , Óxido Nítrico Sintasa de Tipo I/genética , Estado de Hidratación del Organismo , Concentración Osmolar , Factores Sexuales , Transducción de Señal , Urodinámica , Privación de Agua
4.
Am J Physiol Renal Physiol ; 316(5): F875-F888, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30810062

RESUMEN

Deranged histone deacetylase (HDAC) activity causes uncontrolled proliferation, inflammation, fibrosis, and organ damage. It is unclear whether deranged HDAC activity results in acute kidney injury in the renal hypoperfusion model of bilateral ischemia-reperfusion injury (IRI) and whether in vivo inhibition is an appropriate therapeutic approach to limit injury. Male mice were implanted with intraperitoneal osmotic minipumps containing vehicle, the class I HDAC inhibitor, MS275, or the pan-HDAC inhibitor, trichostatin A (TSA), 3 days before sham/bilateral IRI surgery. Kidney cortical samples were analyzed using histological, immunohistochemical, and Western blotting techniques. HDAC-dependent proliferation rate was measured in immortalized rat epithelial cells and primary mouse or human proximal tubule (PT) cells. There were dynamic changes in cortical HDAC localization and abundance following IRI including a fourfold increase in HDAC4 in the PT. HDAC inhibition resulted in a significantly higher plasma creatinine, increased kidney damage, but reduced interstitial fibrosis compared with vehicle-treated IRI mice. HDAC-inhibited mice had reduced interstitial α-smooth muscle actin, fibronectin expression, and Sirius red-positive area, suggesting that IRI activates HDAC-mediated fibrotic pathways. In vivo proliferation of the kidney epithelium was significantly reduced in TSA-treated, but not MS275-treated, IRI mice, suggesting class II HDACs mediate proliferation. Furthermore, HDAC4 activation increased proliferation of human and mouse PTs. Kidney HDACs are activated during IRI with isoform-specific expression patterns. Our data point to mechanisms whereby IRI activates HDACs resulting in fibrotic pathways but also activation of PT proliferation and repair pathways. This study demonstrates the need to develop isoform-selective HDAC inhibitors for the treatment of renal hypoperfusion-induced injury.


Asunto(s)
Lesión Renal Aguda/enzimología , Proliferación Celular , Células Epiteliales/enzimología , Histona Desacetilasas/metabolismo , Túbulos Renales Proximales/enzimología , Daño por Reperfusión/enzimología , Lesión Renal Aguda/tratamiento farmacológico , Lesión Renal Aguda/patología , Animales , Autofagia , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Inhibidores de Histona Desacetilasas/farmacología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/patología , Masculino , Ratones Endogámicos C57BL , Ratas , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/patología , Transducción de Señal , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...