Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 226
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 320(5): F719-F733, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33719576

RESUMEN

Phosphorylation of the thiazide-sensitive NaCl cotransporter (NCC) in the distal convoluted tubule (DCT) is altered rapidly in response to changes in extracellular K+ concentration ([K+]). High extracellular [K+] is believed to activate specific phosphatases to dephosphorylate NCC, thereby reducing its activity. This process is defective in the human disease familial hyperkalemic hypertension, in which extracellular [K+] fails to dephosphorylate NCC, suggesting an interplay between NCC-activating and NCC-inactivating switches. Here, we explored the role of STE20/SPS1-related proline-alanine-rich protein kinase (SPAK) and intracellular Cl- concentration in the rapid effects of extracellular K+ on NCC phosphorylation. SPAK was found to be rapidly dephosphorylated in vitro in human embryonic kidney cells and ex vivo in kidney slices by high [K+]. Acute high-K+ challenge resulted in DCT1-specific SPAK dephosphorylation in vivo and dissolution of SPAK puncta. In line with the postulate of interplay between activating and inactivating switches, we found that the "on" switch, represented by with no lysine kinase 4 (WNK4)-SPAK, must be turned off for rapid NCC dephosphorylation by high [K+]. Longer-term WNK-SPAK-mediated stimulation, however, altered the sensitivity of the system, as it attenuated rapid NCC dephosphorylation due to acute K+ loading. Although blockade of protein phosphatase (PP)1 increased NCC phosphorylation at baseline, neither PP1 nor PP3, singly or in combination, was essential for NCC dephosphorylation. Overall, our data suggest that NCC phosphorylation is regulated by a dynamic equilibrium between activating kinases and inactivating phosphatases, with kinase inactivation playing a key role in the rapid NCC dephosphorylation by high extracellular K+.NEW & NOTEWORTHY Although a great deal is known about mechanisms by which thiazide-sensitive NaCl cotransporter is phosphorylated and activated, much less is known about dephosphorylation. Here, we show that rapid dephosphorylation by high K+ depends on the Cl- sensitivity of with no lysine kinase 4 and the rapid dephosphorylation of STE20/SPS1-related proline-alanine-rich protein kinase, primarily along the early distal convoluted tubule.


Asunto(s)
Cloruros/metabolismo , Túbulos Renales Distales/enzimología , Potasio en la Dieta/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células HEK293 , Humanos , Cinética , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transporte de Proteínas , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
2.
Am J Physiol Renal Physiol ; 319(3): F423-F435, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32657158

RESUMEN

Cre-lox technology has revolutionized research in renal physiology by allowing site-specific genetic recombination in individual nephron segments. The distal convoluted tubule (DCT), consisting of distinct early (DCT1) and late (DCT2) segments, plays a central role in Na+ and K+ homeostasis. The only established Cre line targeting the DCT is Pvalb-Cre, which is limited by noninducibility, activity along DCT1 only, and activity in neurons. Here, we report the characterization of the first Cre line specific to the entire DCT. CRISPR/Cas9 targeting was used to introduce a tamoxifen-inducible IRES-Cre-ERT2 cassette downstream of the coding region of the Slc12a3 gene encoding the NaCl cotransporter (NCC). The resulting Slc12a3-Cre-ERT2 mice were crossed with R26R-YFP reporter mice, which revealed minimal leakiness with 6.3% of NCC-positive cells expressing yellow fluorescent protein (YFP) in the absence of tamoxifen. After tamoxifen injection, YFP expression was observed in 91.2% of NCC-positive cells and only in NCC-positive cells, revealing high recombination efficiency and DCT specificity. Crossing to R26R-TdTomato mice revealed higher leakiness (64.5%), suggesting differential sensitivity of the floxed site. Western blot analysis revealed no differences in abundances of total NCC or the active phosphorylated form of NCC in Slc12a3-Cre-ERT2 mice of either sex compared with controls. Plasma K+ and Mg2+ concentrations and thiazide-sensitive Na+ and K+ excretion did not differ in Slc12a3-Cre-ERT2 mice compared with controls when sex matched. These data suggest genetic modification had no obvious effect on NCC function. Slc12a3-Cre-ERT2 mice are the first line generated demonstrating inducible Cre recombinase activity along the entire DCT and will be a useful tool to study DCT function.


Asunto(s)
Túbulos Renales Distales/enzimología , Recombinasas/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Animales , Antagonistas de Estrógenos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Recombinasas/genética , Simportadores del Cloruro de Sodio/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Tamoxifeno/farmacología
3.
J Hum Hypertens ; 33(7): 508-523, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30723251

RESUMEN

Renal salt handling has a profound effect on body fluid and blood pressure (BP) maintenance as exemplified by the use of diuretic medications to treat states of volume expansion or hypertension. It has recently been proposed that a low potassium (K+) intake turns on a "renal K+ switch" which increases sodium (Na+) and chloride (Cl-) reabsorption, causing salt-retention, and in susceptible individuals, this causes hypertension. A signaling network, involving with-no-lysine (WNK) kinases, underpins the switch activity to coordinate aldosterone's two essential actions (K+ secretion and Na+ retention). A dysfunctional WNK kinase network drives excessive and inappropriate Na+, Cl- and urinary K+ retention in familial hyperkalemic hypertension (FHHt, also known as Gordon's syndrome). Mutations in genes encoding WNK1 and WNK4 or components of an ubiquitin ligase complex, cullin3, and kelch-like family member 3 (KLHL3), cause FHHt by upregulating the thiazide-sensitive sodium chloride cotransporter (NCC). Inhibition of NCC with thiazide diuretics corrects hypertension and hyperkalaemia in FHHt. These observations highlight the critical role of the NCC in the regulation of Na+ and K+ balance and of BP. Here we discuss the physiology of Na+ and K+ handling in the distal renal tubule with respect to BP regulation, with a focus on recent discoveries in the WNK- Ste20-related proline-alanine-rich kinase (SPAK)-NCC pathway.


Asunto(s)
Presión Sanguínea , Hipertensión/enzimología , Túbulos Renales Distales/enzimología , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sodio/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Humanos , Hipertensión/etiología , Hipertensión/fisiopatología , Túbulos Renales Distales/fisiopatología , Masculino , Natriuresis , Reabsorción Renal , Factores de Riesgo , Transducción de Señal , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Equilibrio Hidroelectrolítico
4.
Am J Physiol Renal Physiol ; 316(2): F292-F300, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30484345

RESUMEN

Hypokalemia contributes to the progression of chronic kidney disease, although a definitive pathophysiological theory to explain this remains to be established. K+ deficiency results in profound alterations in renal epithelial transport. These include an increase in salt reabsorption via the Na+-Cl- cotransporter (NCC) of the distal convoluted tubule (DCT), which minimizes electroneutral K+ loss in downstream nephron segments. In experimental conditions of dietary K+ depletion, punctate structures in the DCT containing crucial NCC-regulating kinases have been discovered in the murine DCT and termed "WNK bodies," referring to their component, with no K (lysine) kinases (WNKs). We hypothesized that in humans, WNK bodies occur in hypokalemia as well. Renal needle biopsies of patients with chronic hypokalemic nephropathy and appropriate controls were examined by histological stains and immunofluorescence. Segment- and organelle-specific marker proteins were used to characterize the intrarenal and subcellular distribution of established WNK body constituents, namely, WNKs and Ste20-related proline-alanine-rich kinase (SPAK). In both patients with hypokalemia, WNKs and SPAK concentrated in non-membrane-bound cytoplasmic regions in the DCT, consistent with prior descriptions of WNK bodies. The putative WNK bodies were located in the perinuclear region close to, but not within, the endoplasmic reticulum. They were closely adjacent to microtubules but not clustered in aggresomes. Notably, we provide the first report of WNK bodies, which are functionally challenging structures associated with K+ deficiency, in human patients.


Asunto(s)
Hipopotasemia/enzimología , Enfermedades Renales/enzimología , Túbulos Renales Distales/enzimología , Potasio/sangre , Proteínas Serina-Treonina Quinasas/análisis , Biomarcadores/sangre , Estudios de Casos y Controles , Humanos , Hipopotasemia/sangre , Hipopotasemia/patología , Enfermedades Renales/sangre , Enfermedades Renales/patología , Túbulos Renales Distales/ultraestructura , Complejos Multienzimáticos , Proteína Quinasa Deficiente en Lisina WNK 1/análisis
5.
Am J Physiol Renal Physiol ; 315(3): F429-F444, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29993276

RESUMEN

The vacuolar-type H+-ATPase B1 subunit is heavily expressed in the intercalated cells of the collecting system, where it contributes to H+ transport, but has also been described in other segments of the renal tubule. This study aimed to determine the localization of the B1 subunit of the vacuolar-type H+-ATPase in the early distal nephron, encompassing thick ascending limbs (TAL) and distal convoluted tubules (DCT), in human kidney and determine whether the localization differs between rodents and humans. Antibodies directed against the H+-ATPase B1 subunit were used to determine its localization in paraffin-embedded formalin-fixed mouse, rat, and human kidneys by light microscopy and in sections of Lowicryl-embedded rat kidneys by electron microscopy. Abundant H+-ATPase B1 subunit immunoreactivity was observed in the human kidney. As expected, intercalated cells showed the strongest signal, but significant signal was also observed in apical membrane domains of the distal nephron, including TAL, macula densa, and DCT. In mouse and rat, H+-ATPase B1 subunit expression could also be detected in apical membrane domains of these segments. In rat, electron microscopy revealed that the H+-ATPase B1 subunit was located in the apical membrane. Furthermore, the H+-ATPase B1 subunit colocalized with other H+-ATPase subunits in the TAL and DCT. In conclusion, the B1 subunit is expressed in the early distal nephron. The physiological importance of H+-ATPase expression in these segments remains to be delineated in detail. The phenotype of disease-causing mutations in the B1 subunit may also relate to its presence in the TAL and DCT.


Asunto(s)
Túbulos Renales Distales/enzimología , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Polaridad Celular , Humanos , Inmunohistoquímica , Túbulos Renales Distales/ultraestructura , Ratones Noqueados , Microscopía Electrónica de Transmisión , Especificidad de la Especie , ATPasas de Translocación de Protón Vacuolares/deficiencia , ATPasas de Translocación de Protón Vacuolares/genética
6.
Am J Physiol Renal Physiol ; 315(2): F223-F230, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29667910

RESUMEN

With-no-lysine kinase 4 (WNK4) and kidney-specific (KS)-WNK1 regulate ROMK (Kir1.1) channels in a variety of cell models. We now explore the role of WNK4 and KS-WNK1 in regulating ROMK in the native distal convoluted tubule (DCT)/connecting tubule (CNT) by measuring tertiapin-Q (TPNQ; ROMK inhibitor)-sensitive K+ currents with whole cell recording. TPNQ-sensitive K+ currents in DCT2/CNT of KS- WNK1-/- and WNK4-/- mice were significantly smaller than that of WT mice. In contrast, the basolateral K+ channels (a Kir4.1/5.1 heterotetramer) in the DCT were not inhibited. Moreover, WNK4-/- mice were hypokalemic, while KS- WNK1-/- mice had normal plasma K+ levels. High K+ (HK) intake significantly increased TPNQ-sensitive K+ currents in DCT2/CNT of WT and WNK4-/- mice but not in KS- WNK1-/- mice. However, TPNQ-sensitive K+ currents in the cortical collecting duct (CCD) were normal not only under control conditions but also significantly increased in response to HK in KS- WNK1-/- mice. This suggests that the deletion of KS-WNK1-induced inhibition of ROMK occurs only in the DCT2/CNT. Renal clearance study further demonstrated that the deletion of KS-WNK1 did not affect the renal ability of K+ excretion under control conditions and during increasing K+ intake. Also, HK intake did not cause hyperkalemia in KS- WNK1-/- mice. We conclude that KS-WNK1 but not WNK4 is required for HK intake-induced stimulation of ROMK activity in DCT2/CNT. However, KS-WNK1 is not essential for HK-induced stimulation of ROMK in the CCD, and the lack of KS-WNK1 does not affect net renal K+ excretion.


Asunto(s)
Túbulos Renales Distales/enzimología , Canales de Potasio de Rectificación Interna/metabolismo , Potasio en la Dieta/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Animales , Femenino , Genotipo , Hiperpotasemia/enzimología , Hiperpotasemia/genética , Hipopotasemia/enzimología , Hipopotasemia/genética , Técnicas In Vitro , Masculino , Potenciales de la Membrana , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Canales de Potasio de Rectificación Interna/genética , Potasio en la Dieta/orina , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Eliminación Renal , Proteína Quinasa Deficiente en Lisina WNK 1/deficiencia , Proteína Quinasa Deficiente en Lisina WNK 1/genética
7.
Am J Physiol Renal Physiol ; 314(5): F999-F1007, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29384416

RESUMEN

The Na+-Cl- cotransporter (NCC) in distal convoluted tubule (DCT) plays important roles in renal NaCl reabsorption. The current hypothesis for the mechanism of regulation of NCC focuses on WNK4 and intracellular Cl- concentration ([Cl-]i). WNK kinases bind Cl-, and Cl- binding decreases the catalytic activity. It is believed that hypokalemia under low K+ intake decreases [Cl-]i to activate WNK4, which thereby phosphorylates and stimulates NCC through activation of SPAK. However, increased NCC activity and apical NaCl entry would mitigate the fall in [Cl-]i. Whether [Cl-]i in DCT under low-K+ diet is sufficiently low to activate WNK4 is unknown. Furthermore, increased luminal NaCl delivery also stimulates NCC and causes upregulation of the transporter. Unlike low K+ intake, increased luminal NaCl delivery would tend to increase [Cl-]i. Thus we investigated the role of WNK4 and [Cl-]i in regulating NCC. We generated Wnk4-knockout mice and examined regulation of NCC by low K+ intake and by increased luminal NaCl delivery in knockout (KO) and wild-type mice. Wnk4-KO mice have marked reduction in the abundance, phosphorylation, and functional activity of NCC vs. wild type. Low K+ intake increases NCC phosphorylation and functional activity in wild-type mice, but not in Wnk4-KO mice. Increased luminal NaCl delivery similarly upregulates NCC, which, contrary to low K+ intake, is not abolished in Wnk4-KO mice. The results reveal that modulation of WNK4 activity by [Cl-]i is not the sole mechanism for regulating NCC. Increased luminal NaCl delivery upregulates NCC via yet unknown mechanism(s) that may override inhibition of WNK4 by high [Cl-]i.


Asunto(s)
Túbulos Renales Distales/enzimología , Potasio en la Dieta/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Cloruro de Sodio/metabolismo , Animales , Transporte Biológico , Regulación Enzimológica de la Expresión Génica , Inyecciones Subcutáneas , Túbulos Renales Distales/efectos de los fármacos , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Eliminación Renal , Reabsorción Renal , Cloruro de Sodio/administración & dosificación , Inhibidores de los Simportadores del Cloruro de Sodio/farmacología , Miembro 3 de la Familia de Transportadores de Soluto 12/deficiencia , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
8.
J Am Soc Nephrol ; 28(8): 2431-2442, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28289184

RESUMEN

Adaptation of the organism to potassium (K+) deficiency requires precise coordination among organs involved in K+ homeostasis, including muscle, liver, and kidney. How the latter performs functional and molecular changes to ensure K+ retention is not well understood. Here, we investigated the role of ubiquitin-protein ligase NEDD4-2, which negatively regulates the epithelial sodium channel (ENaC), Na+/Cl- cotransporter (NCC), and with no-lysine-kinase 1 (WNK1). After dietary K+ restriction for 2 weeks, compared with control littermates, inducible renal tubular NEDD4-2 knockout (Nedd4LPax8/LC1 ) mice exhibited severe hypokalemia and urinary K+ wasting. Notably, expression of the ROMK K+ channel did not change in the distal convoluted tubule and decreased slightly in the cortical/medullary collecting duct, whereas BK channel abundance increased in principal cells of the connecting tubule/collecting ducts. However, K+ restriction also enhanced ENaC expression in Nedd4LPax8/LC1 mice, and treatment with the ENaC inhibitor, benzamil, reversed excessive K+ wasting. Moreover, K+ restriction increased WNK1 and WNK4 expression and enhanced SPAK-mediated NCC phosphorylation in Nedd4LPax8/LC1 mice, with no change in total NCC. We propose a mechanism in which NEDD4-2 deficiency exacerbates hypokalemia during dietary K+ restriction primarily through direct upregulation of ENaC, whereas increased BK channel expression has a less significant role. These changes outweigh the compensatory antikaliuretic effects of diminished ROMK expression, increased NCC phosphorylation, and enhanced WNK pathway activity in the distal convoluted tubule. Thus, NEDD4-2 has a crucial role in K+ conservation through direct and indirect effects on ENaC, distal nephron K+ channels, and WNK signaling.


Asunto(s)
Adaptación Fisiológica , Complejos de Clasificación Endosomal Requeridos para el Transporte/fisiología , Hipopotasemia/fisiopatología , Túbulos Renales Distales/enzimología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Riñón/fisiopatología , Ratones , Ubiquitina-Proteína Ligasas Nedd4 , Factores de Tiempo
9.
Curr Res Transl Med ; 64(1): 5-8, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27140593

RESUMEN

BACKGROUND: Primary distal renal tubular acidosis (dRTA) is a rare genetic condition characterized by an impaired acid excretion by the intercalated cells in the renal collecting duct. Recessive forms of this disease are caused by mutations in tow major genes: ATP6V1B1 and ATP6V0A4. Causal mutations in ATP6V1B1 gene are classically associated with early sensorineural hearing loss, however cases of tubular acidosis with early deafness have also been described in patients with mutations in the ATP6V0A4 gene. METHODS: The phenotype and genotype of three Moroccan consanguineous families with dRTA and deafness were assessed. Molecular analysis was performed by PCR amplification and direct sequencing of exon 12 of ATP6V1B1 gene. RESULTS: A novel c.1169dupC frameshift mutation of ATP6V1B1 gene was identified in one family and the c.1155dupC North African mutation in the tow other families. DISCUSSION AND CONCLUSION: In this report, we propose first line genetic testing based on screening of these two mutations both located in exon 12 of ATP6V1B1 gene in Moroccan patients with recessive form of dRTA associated to precocious hearing loss. Molecular diagnosis of dRTA leads to appropriate treatment and prevention of renal failure in affected individuals and to provide genetic counseling for families at risk.


Asunto(s)
Acidosis Tubular Renal/genética , Codón sin Sentido , Sordera/genética , Mutación del Sistema de Lectura , Pérdida Auditiva Sensorineural/genética , ATPasas de Translocación de Protón Vacuolares/genética , Edad de Inicio , Sustitución de Aminoácidos , Cóclea/enzimología , Consanguinidad , Diagnóstico Precoz , Exones/genética , Femenino , Genes Recesivos , Humanos , Lactante , Túbulos Renales Distales/enzimología , Masculino , Marruecos , Nefrocalcinosis/genética , Linaje , ATPasas de Translocación de Protón Vacuolares/deficiencia
10.
PLoS One ; 11(1): e0147785, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26820468

RESUMEN

Group V (GV) phospholipase A2 (PLA2) is a member of the family of secreted PLA2 (sPLA2) enzymes. This enzyme has been identified in several organs, including the kidney. However, the physiologic role of GV sPLA2 in the maintenance of renal function remains unclear. We used mice lacking the gene encoding GV sPLA2 (Pla2g5-/-) and wild-type breeding pairs in the experiments. Mice were individually housed in metabolic cages and 48-h urine was collected for biochemical assays. Kidney samples were evaluated for glomerular morphology, renal fibrosis, and expression/activity of the (Na+ + K+)-ATPase α1 subunit. We observed that plasma creatinine levels were increased in Pla2g5-/- mice following by a decrease in creatinine clearance. The levels of urinary protein were higher in Pla2g5-/- mice than in the control group. Markers of tubular integrity and function such as γ-glutamyl transpeptidase, lactate dehydrogenase, and sodium excretion fraction (FENa+) were also increased in Pla2g5-/- mice. The increased FENa+ observed in Pla2g5-/- mice was correlated to alterations in cortical (Na+ + K+) ATPase activity/ expression. In addition, the kidney from Pla2g5-/- mice showed accumulation of matrix in corticomedullary glomeruli and tubulointerstitial fibrosis. These data suggest GV sPLA2 is involved in the maintenance of tubular cell function and integrity, promoting sodium retention through increased cortical (Na+ + K+)-ATPase expression and activity.


Asunto(s)
Fosfolipasas A2 Grupo V/fisiología , Túbulos Renales Distales/enzimología , Riñón/enzimología , Sodio/metabolismo , Animales , Homeostasis , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , ATPasa Intercambiadora de Sodio-Potasio/metabolismo
11.
Am J Physiol Renal Physiol ; 308(8): F923-31, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25651563

RESUMEN

The NaCl cotransporter (NCC) of the renal distal convoluted tubule is stimulated by low-K(+) diet by an unknown mechanism. Since recent work has shown that the STE20/SPS-1-related proline-alanine-rich protein kinase (SPAK) can function to stimulate NCC by phosphorylation of specific N-terminal sites, we investigated whether the NCC response to low-K(+) diet is mediated by SPAK. Using phospho-specific antibodies in Western blot and immunolocalization studies of wild-type and SPAK knockout (SPAK(-/-)) mice fed a low-K(+) or control diet for 4 days, we found that low-K(+) diet strongly increased total NCC expression and phosphorylation of NCC. This was associated with an increase in total SPAK expression in cortical homogenates and an increase in phosphorylation of SPAK at the S383 activation site. The increased pNCC in response to low-K(+) diet was blunted but not completely inhibited in SPAK(-/-) mice. These findings reveal that SPAK is an important mediator of the increased NCC activation by phosphorylation that occurs in the distal convoluted tubule in response to a low-K(+) diet, but other low-potassium-activated kinases are likely to be involved.


Asunto(s)
Túbulos Renales Distales/enzimología , Deficiencia de Potasio/enzimología , Potasio en la Dieta/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Ratones Noqueados , Fosforilación , Deficiencia de Potasio/genética , Potasio en la Dieta/administración & dosificación , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Transducción de Señal , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Regulación hacia Arriba
12.
Am J Physiol Cell Physiol ; 307(6): C532-41, 2014 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-25031022

RESUMEN

Cumulative evidence suggests that guanylin peptides play an important role on electrolyte homeostasis. We have previously reported that uroguanylin (UGN) inhibits bicarbonate reabsorption in a renal distal tubule. In the present study, we tested the hypothesis that the bicarbonaturic effect of UGN is at least in part attributable to inhibition of H(+)-ATPase-mediated hydrogen secretion in the distal nephron. By in vivo stationary microperfusion experiments, we were able to show that UGN inhibits H(+)-ATPase activity by a PKG-dependent pathway because KT5823 (PKG inhibitor) abolished the UGN effect on distal bicarbonate reabsorption and H89 (PKA inhibitor) was unable to prevent it. The in vivo results were confirmed by the in vitro experiments, where we used fluorescence microscopy to measure intracellular pH (pHi) recovery after an acid pulse with NH4Cl. By this technique, we observed that UGN and 8 bromoguanosine-cGMP (8Br-cGMP) inhibited H(+)-ATPase-dependent pHi recovery and that the UGN inhibitory effect was abolished in the presence of the PKG inhibitor. In addition, by using RT-PCR technique, we verified that Madin-Darby canine kidney (MDCK)-C11 cells express guanylate cyclase-C. Besides, UGN stimulated an increase of both cGMP content and PKG activity but was unable to increase the production of cellular cAMP content and PKA activity. Furthermore, we found that UGN reduced cell surface abundance of H+-ATPase B1 subunit in MDCK-C11 and that this effect was abolished by the PKG inhibitor. Taken together, our data suggest that UGN inhibits H(+)-ATPase activity and surface expression in renal distal cells by a cGMP/PKG-dependent pathway.


Asunto(s)
Membrana Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Túbulos Renales Distales/efectos de los fármacos , Péptidos Natriuréticos/farmacología , ATPasas de Translocación de Protón/metabolismo , Animales , Bicarbonatos/metabolismo , Membrana Celular/enzimología , GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/antagonistas & inhibidores , Perros , Concentración de Iones de Hidrógeno , Túbulos Renales Distales/enzimología , Células de Riñón Canino Madin Darby , Masculino , Perfusión , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas , Ratas , Ratas Wistar , Receptores Acoplados a la Guanilato-Ciclasa/efectos de los fármacos , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
13.
J Biol Chem ; 289(17): 11791-11806, 2014 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-24610784

RESUMEN

It has been well established that blood pressure and renal function undergo circadian fluctuations. We have demonstrated that the circadian protein Per1 regulates multiple genes involved in sodium transport in the collecting duct of the kidney. However, the role of Per1 in other parts of the nephron has not been investigated. The distal convoluted tubule (DCT) plays a critical role in renal sodium reabsorption. Sodium is reabsorbed in this segment through the actions of the NaCl co-transporter (NCC), which is regulated by the with-no-lysine kinases (WNKs). The goal of this study was to test if Per1 regulates sodium transport in the DCT through modulation of NCC and the WNK kinases, WNK1 and WNK4. Pharmacological blockade of nuclear Per1 entry resulted in decreased mRNA expression of NCC and WNK1 but increased expression of WNK4 in the renal cortex of mice. These findings were confirmed by using Per1 siRNA and pharmacological blockade of Per1 nuclear entry in mDCT15 cells, a model of the mouse distal convoluted tubule. Transcriptional regulation was demonstrated by changes in short lived heterogeneous nuclear RNA. Chromatin immunoprecipitation experiments demonstrated interaction of Per1 and CLOCK with the promoters of NCC, WNK1, and WNK4. This interaction was modulated by blockade of Per1 nuclear entry. Importantly, NCC protein expression and NCC activity, as measured by thiazide-sensitive, chloride-dependent (22)Na uptake, were decreased upon pharmacological inhibition of Per1 nuclear entry. Taken together, these data demonstrate a role for Per1 in the transcriptional regulation of NCC, WNK1, and WNK4.


Asunto(s)
Túbulos Renales Distales/metabolismo , Proteínas Circadianas Period/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Núcleo Celular/metabolismo , Inmunoprecipitación de Cromatina , Cartilla de ADN , Técnicas de Silenciamiento del Gen , Túbulos Renales Distales/enzimología , Ratones , Ratones Noqueados , Proteínas Circadianas Period/genética , Proteínas Serina-Treonina Quinasas/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
14.
BMC Res Notes ; 7: 116, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24576305

RESUMEN

BACKGROUND: Carbonic anhydrase VI (CA-VI) is produced by the salivary gland and is secreted into the saliva. Although CA-VI is found in the epithelial cells of distal straight tubule of swine kidneys, the exact function of CA-VI in the kidneys remains unclear. RESULTS: CA-VI was located in the epithelial cells of distal straight tubule of swine kidneys.A full-length cDNA clone of CA-VI was generated from the swine parotid gland by reverse transcription polymerase chain reaction, using degenerate primers designed based on conserved regions of the same locus in human and bovine tissues. The cDNA sequence was 1348 base pairs long and was predicted to encode a 317 amino acid polypeptide with a putative signal peptide of 17 amino acids. The deduced amino acid sequence of mature CA-VI was most similar (77.4%) to that of human CA-VI. CA-VI expression was confirmed in both normal and nephritic kidneys, as well as parotid. As the primers used in this study spanned two exons, the influence of genomic DNA was not detected. The expression of CA-VI was demonstrated in both normal and nephritic kidneys, and mRNA of CA-VI in the normal kidneys which was the normalised to an endogenous ß-actin was 0.098 ± 0.047, while it was significantly lower in the diseased kidneys (0.012 ± 0.007). The level of CA-VI mRNA in normal kidneys was 19-fold lower than that of the parotid gland (1.887). CONCLUSIONS: The localisation of CA-VI indicates that it may play a specialised role in the kidney.


Asunto(s)
Anhidrasas Carbónicas/genética , Células Epiteliales/metabolismo , Regulación Enzimológica de la Expresión Génica , Túbulos Renales Distales/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Anhidrasas Carbónicas/aislamiento & purificación , Anhidrasas Carbónicas/metabolismo , Clonación Molecular , ADN Complementario/química , ADN Complementario/genética , Células Epiteliales/enzimología , Inmunohistoquímica , Túbulos Renales Distales/citología , Túbulos Renales Distales/enzimología , Datos de Secuencia Molecular , Nefritis/enzimología , Nefritis/genética , Glándula Parótida/enzimología , Glándula Parótida/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Secuencia de ADN , Homología de Secuencia de Aminoácido , Porcinos
15.
J Am Soc Nephrol ; 25(3): 511-22, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24231659

RESUMEN

The thiazide-sensitive NaCl cotransporter (NCC) of the renal distal convoluted tubule (DCT) controls ion homeostasis and arterial BP. Loss-of-function mutations of NCC cause renal salt wasting with arterial hypotension (Gitelman syndrome). Conversely, mutations in the NCC-regulating WNK kinases or kelch-like 3 protein cause familial hyperkalemic hypertension. Here, we performed automated sorting of mouse DCTs and microarray analysis for comprehensive identification of novel DCT-enriched gene products, which may potentially regulate DCT and NCC function. This approach identified protein phosphatase 1 inhibitor-1 (I-1) as a DCT-enriched transcript, and immunohistochemistry revealed I-1 expression in mouse and human DCTs and thick ascending limbs. In heterologous expression systems, coexpression of NCC with I-1 increased thiazide-dependent Na(+) uptake, whereas RNAi-mediated knockdown of endogenous I-1 reduced NCC phosphorylation. Likewise, levels of phosphorylated NCC decreased by approximately 50% in I-1 (I-1(-/-)) knockout mice without changes in total NCC expression. The abundance and phosphorylation of other renal sodium-transporting proteins, including NaPi-IIa, NKCC2, and ENaC, did not change, although the abundance of pendrin increased in these mice. The abundance, phosphorylation, and subcellular localization of SPAK were similar in wild-type (WT) and I-1(-/-) mice. Compared with WT mice, I-1(-/-) mice exhibited significantly lower arterial BP but did not display other metabolic features of NCC dysregulation. Thus, I-1 is a DCT-enriched gene product that controls arterial BP, possibly through regulation of NCC activity.


Asunto(s)
Hipotensión/enzimología , Túbulos Renales Distales/enzimología , Proteínas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Proteínas de Transporte de Anión/metabolismo , Presión Sanguínea , Femenino , Humanos , Asa de la Nefrona/enzimología , Masculino , Ratones , Ratones Transgénicos , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/genética , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Transportadores de Sulfato , Regulación hacia Arriba , Xenopus
16.
Shock ; 41(3): 256-65, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24296430

RESUMEN

In septic shock (SS), dysfunction of many organ systems develops during the course of the illness, although the mechanisms are not clear. In earlier studies, we reported that lysozyme-c (Lzm-S), a protein that is released from leukocytes and macrophages, was a mediator of the myocardial depression and vasodilation that develop in a canine model of Pseudomonas aeruginosa SS. Whereas both of these effects of Lzm-S are dependent on its ability to intrinsically generate hydrogen peroxide, we subsequently showed that Lzm-S can also deposit within the vascular smooth muscle layer of the systemic arteries in this model. In the present study, we extend our previous findings. We used a canine carotid artery organ bath preparation to study the time course and dose dependence of Lzm-S deposition within the vascular smooth muscle layer. We used a human aortic vascular smooth muscle cell preparation to determine whether Lzm-S can persistently inhibit contraction in this preparation. We also used a canine P. aeruginosa model to determine whether Lzm-S deposition might occur in other organs such as the kidney, liver, and small intestine. The results showed that, in the carotid artery organ bath preparation, Lzm-S deposition occurred within minutes of instillation and there was a dose-response effect. In the human aortic vascular smooth muscle cell preparation, Lzm-S inhibited contraction during a 4-day period. In the in vivo model, Lzm-S accumulated in the kidney and the superior mesenteric artery. In a canine renal epithelial preparation, we further showed that Lzm-S can be taken up by the renal tubules to activate inflammatory pathways. We conclude that Lzm-S can deposit in the systemic vasculature and kidneys in SS, where this deposition could lead to acute organ dysfunction.


Asunto(s)
Túbulos Renales Distales/enzimología , Macrófagos/enzimología , Muramidasa/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Sepsis/enzimología , Animales , Aorta/enzimología , Aorta/patología , Arterias Carótidas/enzimología , Arterias Carótidas/patología , Células Cultivadas , Modelos Animales de Enfermedad , Perros , Humanos , Intestino Delgado/enzimología , Intestino Delgado/patología , Túbulos Renales Distales/patología , Arterias Mesentéricas/enzimología , Arterias Mesentéricas/patología , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Técnicas de Cultivo de Órganos , Infecciones por Pseudomonas/enzimología , Infecciones por Pseudomonas/patología , Pseudomonas aeruginosa , Sepsis/patología
17.
Am J Physiol Renal Physiol ; 305(7): F995-F1005, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23863470

RESUMEN

Nadph oxidase 4 is an important cellular source of reactive oxygen species (ROS) generation in the kidney. Novel antioxidant drugs, such as Nox4 inhibitor compounds, are being developed. There is, however, very little experimental evidence for the biological role and regulation of Nadph oxidase isoforms in the kidney. Herein, we show that Fulvene-5 is an effective inhibitor of Nox-generated ROS and report the role of Nox isoforms in activating epithelial sodium channels (ENaC) in A6 distal nephron cells via oxidant signaling and cell stretch activation. Using single-channel patch-clamp analysis, we report that Fulvene-5 blocked the increase in ENaC activity that is typically observed with H2O2 treatment of A6 cells: average ENaC NPo values decreased from a baseline level of 1.04 ± 0.18 (means ± SE) to 0.25 ± 0.08 following Fulvene-5 treatment. H2O2 treatment failed to increase ENaC activity in the presence of Fulvene-5. Moreover, Fulvene-5 treatment of A6 cells blocked the osmotic cell stretch response of A6 cells, indicating that stretch activation of Nox-derived ROS plays an important role in ENaC regulation. Together, these findings indicate that Fulvene-5, and perhaps other classes of antioxidant inhibitors, may represent a novel class of compounds useful for the treatment of pathological disorders stemming from inappropriate ion channel activity, such as hypertension.


Asunto(s)
Ciclopentanos/farmacología , Canales Epiteliales de Sodio/metabolismo , Túbulos Renales Distales/enzimología , NADPH Oxidasas/antagonistas & inhibidores , NADPH Oxidasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Línea Celular , Túbulos Renales Distales/citología , Túbulos Renales Distales/efectos de los fármacos , Ósmosis/efectos de los fármacos , Xenopus
18.
PLoS One ; 6(8): e24277, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21909387

RESUMEN

The NaCl cotransporter (NCC) is essential for sodium reabsorption at the distal convoluted tubules (DCT), and its phosphorylation increases its transport activity and apical membrane localization. Although insulin has been reported to increase sodium reabsorption in the kidney, the linkage between insulin and NCC phosphorylation has not yet been investigated. This study examined whether insulin regulates NCC phosphorylation. In cultured mpkDCT cells, insulin increased phosphorylation of STE20/SPS1-related proline-alanine-rich kinase (SPAK) and NCC in a dose-dependent manner. This insulin-induced phosphorylation of NCC was suppressed in WNK4 and SPAK knockdown cells. In addition, Ly294002, a PI3K inhibitor, decreased the insulin effect on SPAK and NCC phosphorylation, indicating that insulin induces phosphorylation of SPAK and NCC through PI3K and WNK4 in mpkDCT cells. Moreover, acute insulin administration to mice increased phosphorylation of oxidative stress-responsive kinase-1 (OSR1), SPAK and NCC in the kidney. Time-course experiments in mpkDCT cells and mice suggested that SPAK is upstream of NCC in this insulin-induced NCC phosphorylation mechanism, which was confirmed by the lack of insulin-induced NCC phosphorylation in SPAK knockout mice. Moreover, insulin administration to WNK4 hypomorphic mice did not increase phosphorylation of OSR1, SPAK and NCC in the kidney, suggesting that WNK4 is also involved in the insulin-induced OSR1, SPAK and NCC phosphorylation mechanism in vivo. The present results demonstrated that insulin is a potent regulator of NCC phosphorylation in the kidney, and that WNK4 and SPAK are involved in this mechanism of NCC phosphorylation by insulin.


Asunto(s)
Insulina/farmacología , Túbulos Renales Distales/citología , Túbulos Renales Distales/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Animales , Células Cultivadas , Técnicas de Silenciamiento del Gen , Insulina/administración & dosificación , Túbulos Renales Distales/enzimología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Cloruro de Sodio Dietético/farmacología , Factores de Tiempo
19.
J Am Soc Nephrol ; 22(9): 1707-19, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21852580

RESUMEN

Regulation of renal Na(+) transport is essential for controlling blood pressure, as well as Na(+) and K(+) homeostasis. Aldosterone stimulates Na(+) reabsorption by the Na(+)-Cl(-) cotransporter (NCC) in the distal convoluted tubule (DCT) and by the epithelial Na(+) channel (ENaC) in the late DCT, connecting tubule, and collecting duct. Aldosterone increases ENaC expression by inhibiting the channel's ubiquitylation and degradation; aldosterone promotes serum-glucocorticoid-regulated kinase SGK1-mediated phosphorylation of the ubiquitin-protein ligase Nedd4-2 on serine 328, which prevents the Nedd4-2/ENaC interaction. It is important to note that aldosterone increases NCC protein expression by an unknown post-translational mechanism. Here, we present evidence that Nedd4-2 coimmunoprecipitated with NCC and stimulated NCC ubiquitylation at the surface of transfected HEK293 cells. In Xenopus laevis oocytes, coexpression of NCC with wild-type Nedd4-2, but not its catalytically inactive mutant, strongly decreased NCC activity and surface expression. SGK1 prevented this inhibition in a kinase-dependent manner. Furthermore, deficiency of Nedd4-2 in the renal tubules of mice and in cultured mDCT(15) cells upregulated NCC. In contrast to ENaC, Nedd4-2-mediated inhibition of NCC did not require the PY-like motif of NCC. Moreover, the mutation of Nedd4-2 at either serine 328 or 222 did not affect SGK1 action, and mutation at both sites enhanced Nedd4-2 activity and abolished SGK1-dependent inhibition. Taken together, these results suggest that aldosterone modulates NCC protein expression via a pathway involving SGK1 and Nedd4-2 and provides an explanation for the well-known aldosterone-induced increase in NCC protein expression.


Asunto(s)
Aldosterona/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Proteínas Inmediatas-Precoces/metabolismo , Túbulos Renales Distales/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , 11-beta-Hidroxiesteroide Deshidrogenasa de Tipo 2/metabolismo , Animales , Regulación hacia Abajo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4 , Fosforilación , Transducción de Señal , Ubiquitinación , Proteínas de Xenopus , Xenopus laevis
20.
Histochem Cell Biol ; 136(1): 25-35, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21660484

RESUMEN

Initial reports claim that WNK4 localization is mainly at intercellular junctions of distal convoluted tubules (DCT) and cortical collecting ducts (CCD) in the kidney. However, we recently clarified the major targets of WNK4 kinase to be the OSR1/SPAK kinases and the Na-Cl co-transporter (NCC), an apical membrane protein in the DCT, thus raising the question of whether the cellular localization of WNK4 is at intercellular junctions. In this study, we re-evaluate the intrarenal and intracellular immunolocalization of WNK4 in the mouse kidney using a newly generated anti-WNK4 antibody. By performing double immunofluorescence of WNK4 with several nephron-segment-specific markers, we have found that WNK4 is present in podocytes in glomeruli, the cortical thick ascending limb of Henle's loop including macula densa, and the medullary collecting ducts (MCD), in addition to the previously identified nephron segments, i.e., DCT and CCD. These results are consistent with the finding that WNK4 constitutes a kinase cascade with OSR1/SPAK and NCC in the DCT, and highlights a novel role for WNK4 in nephron segments newly identified as being WNK4-positive in this study.


Asunto(s)
Riñón/enzimología , Proteínas Serina-Treonina Quinasas/análisis , Animales , Células COS , Chlorocebus aethiops , Técnica del Anticuerpo Fluorescente , Riñón/metabolismo , Túbulos Renales Distales/enzimología , Túbulos Renales Distales/metabolismo , Ratones , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Droga/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...