Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 114
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Renal Physiol ; 320(5): F734-F747, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33682442

RESUMEN

The physiological role of the shorter isoform of with no lysine kinase (WNK)1 that is exclusively expressed in the kidney (KS-WNK1), with particular abundance in the distal convoluted tubule, remains elusive. KS-WNK1, despite lacking the kinase domain, is nevertheless capable of stimulating the NaCl cotransporter, apparently through activation of WNK4. It has recently been shown that a less severe form of familial hyperkalemic hypertension featuring only hyperkalemia is caused by missense mutations in the WNK1 acidic domain that preferentially affect cullin 3 (CUL3)-Kelch-like protein 3 (KLHL3) E3-induced degradation of KS-WNK1 rather than that of full-length WNK1. Here, we show that full-length WNK1 is indeed less impacted by the CUL3-KLHL3 E3 ligase complex compared with KS-WNK1. We demonstrated that the unique 30-amino acid NH2-terminal fragment of KS-WNK1 is essential for its activating effect on the NaCl cotransporter and recognition by KLHL3. We identified specific amino acid residues in this region critical for the functional effect of KS-WNK1 and KLHL3 sensitivity. To further explore this, we generated KLHL3-R528H knockin mice that mimic human mutations causing familial hyperkalemic hypertension. These mice revealed that the KLHL3 mutation specifically increased expression of KS-WNK1 in the kidney. We also observed that in wild-type mice, the expression of KS-WNK1 was only detectable after exposure to a low-K+ diet. These findings provide new insights into the regulation and function of KS-WNK1 by the CUL3-KLHL3 complex in the distal convoluted tubule and indicate that this pathway is regulated by dietary K+ levels.NEW & NOTEWORTHY In this work, we demonstrated that the kidney-specific isoform of with no lysine kinase 1 (KS-WNK1) in the kidney is modulated by dietary K+ and activity of the ubiquitin ligase protein Kelch-like protein 3. We analyzed the role of different amino acid residues of KS-WNK1 in its activity against the NaCl cotransporter and sensitivity to Kelch-like protein 3.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Riñón/enzimología , Proteínas de Microfilamentos/metabolismo , Potasio en la Dieta/metabolismo , Seudohipoaldosteronismo/enzimología , Proteína Quinasa Deficiente en Lisina WNK 1/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Cullin/metabolismo , Estabilidad de Enzimas , Femenino , Riñón/fisiopatología , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Mutación , Dominios y Motivos de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteolisis , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1/deficiencia , Proteína Quinasa Deficiente en Lisina WNK 1/genética , Xenopus laevis
2.
Annu Rev Physiol ; 78: 367-89, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26863326

RESUMEN

The discovery of four genes responsible for pseudohypoaldosteronism type II, or familial hyperkalemic hypertension, which features arterial hypertension with hyperkalemia and metabolic acidosis, unmasked a complex multiprotein system that regulates electrolyte transport in the distal nephron. Two of these genes encode the serine-threonine kinases WNK1 and WNK4. The other two genes [kelch-like 3 (KLHL3) and cullin 3 (CUL3)] form a RING-type E3-ubiquitin ligase complex that modulates WNK1 and WNK4 abundance. WNKs regulate the activity of the Na(+):Cl(-) cotransporter (NCC), the epithelial sodium channel (ENaC), the renal outer medullary potassium channel (ROMK), and other transport pathways. Interestingly, the modulation of NCC occurs via the phosphorylation by WNKs of other serine-threonine kinases known as SPAK-OSR1. In contrast, the process of regulating the channels is independent of SPAK-OSR1. We present a review of the remarkable advances in this area in the past 10 years.


Asunto(s)
Electrólitos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transporte Iónico/fisiología , Riñón/metabolismo , Riñón/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Humanos , Hipertensión/metabolismo , Hipertensión/fisiopatología , Seudohipoaldosteronismo/metabolismo , Seudohipoaldosteronismo/fisiopatología
3.
Curr Opin Nephrol Hypertens ; 28(5): 490-497, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31313673

RESUMEN

PURPOSE OF REVIEW: Members of the Cullin family act as scaffolds in E3 ubiquitin ligases and play a central role in mediating protein degradation. Interactions with many different substrate-binding adaptors permit Cullin-containing E3 ligases to participate in diverse cellular functions. In the kidney, one well established target of Cullin-mediated degradation is the transcription factor Nrf2, a key player in responses to oxidative stress. The goal of this review is to discuss more recent findings revealing broader roles for Cullins in the kidney. RECENT FINDINGS: Cullin 3 acts as the scaffold in the E3 ligase regulating Nrf2 abundance, but was more recently shown to be mutated in the disease familial hyperkalemic hypertension. Studies seeking to elucidate the molecular mechanisms by which Cullin 3 mutations lead to dysregulation of renal sodium transport will be discussed. Disruption of Cullin 3 in mice unexpectedly causes polyuria and fibrotic injury suggesting it has additional roles in the kidney. We will also review recent transcriptomic data suggesting that other Cullins are also likely to play important roles in renal function. SUMMARY: Cullins form a large and diverse family of E3 ubiquitin ligases that are likely to have many important functions in the kidney.


Asunto(s)
Proteínas Cullin/fisiología , Enfermedades Renales/etiología , Riñón/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Carcinoma de Células Renales/etiología , Humanos , Neoplasias Renales/etiología , Proteínas de Microfilamentos/fisiología , Factor 2 Relacionado con NF-E2/fisiología , Seudohipoaldosteronismo/etiología , Seudohipoaldosteronismo/fisiopatología , Simportadores del Cloruro de Sodio/fisiología
4.
Am J Physiol Renal Physiol ; 314(5): F915-F920, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29361671

RESUMEN

Autosomal dominant mutations in cullin-3 ( Cul3) cause the most severe form of familial hyperkalemic hypertension (FHHt). Cul3 mutations cause skipping of exon 9, which results in an internal deletion of 57 amino acids from the CUL3 protein (CUL3-∆9). The precise mechanism by which this altered form of CUL3 causes FHHt is controversial. CUL3 is a member of the cullin-RING ubiquitin ligase family that mediates ubiquitination and thus degradation of cellular proteins, including with-no-lysine [K] kinases (WNKs). In CUL3-∆9-mediated FHHt, proteasomal degradation of WNKs is abrogated, leading to overactivation of the WNK targets sterile 20/SPS-1 related proline/alanine-rich kinase and oxidative stress-response kinase-1, which directly phosphorylate and activate the thiazide-sensitive Na+-Cl- cotransporter. Several groups have suggested different mechanisms by which CUL3-∆9 causes FHHt. The majority of these are derived from in vitro data, but recently the Kurz group (Schumacher FR, Siew K, Zhang J, Johnson C, Wood N, Cleary SE, Al Maskari RS, Ferryman JT, Hardege I, Figg NL, Enchev R, Knebel A, O'Shaughnessy KM, Kurz T. EMBO Mol Med 7: 1285-1306, 2015) described the first mouse model of CUL3-∆9-mediated FHHt. Analysis of this model suggested that CUL3-∆9 is degraded in vivo, and thus Cul3 mutations cause FHHt by inducing haploinsufficiency. We recently directly tested this model but found that other dominant effects of CUL3-∆9 must contribute to the development of FHHt. In this review, we focus on our current knowledge of CUL3-∆9 action gained from in vitro and in vivo models that may help unravel this complex problem.


Asunto(s)
Presión Sanguínea , Proteínas Cullin/genética , Mutación , Nefronas/enzimología , Seudohipoaldosteronismo/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Presión Sanguínea/genética , Proteínas Portadoras/metabolismo , Proteínas Cullin/metabolismo , Modelos Animales de Enfermedad , Estabilidad de Enzimas , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Humanos , Proteínas de Microfilamentos , Nefronas/fisiopatología , Fenotipo , Seudohipoaldosteronismo/diagnóstico , Seudohipoaldosteronismo/enzimología , Seudohipoaldosteronismo/fisiopatología
5.
Kidney Int ; 94(3): 514-523, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30146013

RESUMEN

Pseudohypoaldosteronism type II (PHAII) is a genetic disease characterized by association of hyperkalemia, hyperchloremic metabolic acidosis, hypertension, low renin, and high sensitivity to thiazide diuretics. It is caused by mutations in the WNK1, WNK4, KLHL3 or CUL3 gene. There is strong evidence that excessive sodium chloride reabsorption by the sodium chloride cotransporter NCC in the distal convoluted tubule is involved. WNK4 is expressed not only in distal convoluted tubule cells but also in ß-intercalated cells of the cortical collecting duct. These latter cells exchange intracellular bicarbonate for external chloride through pendrin, and therefore, account for renal base excretion. However, these cells can also mediate thiazide-sensitive sodium chloride absorption when the pendrin-dependent apical chloride influx is coupled to apical sodium influx by the sodium-driven chloride/bicarbonate exchanger. Here we determine whether this system is involved in the pathogenesis of PHAII. Renal pendrin activity was markedly increased in a mouse model carrying a WNK4 missense mutation (Q562E) previously identified in patients with PHAII. The upregulation of pendrin led to an increase in thiazide-sensitive sodium chloride absorption by the cortical collecting duct, and it caused metabolic acidosis. The function of apical potassium channels was altered in this model, and hyperkalemia was fully corrected by pendrin genetic ablation. Thus, we demonstrate an important contribution of pendrin in renal regulation of sodium chloride, potassium and acid-base homeostasis and in the pathophysiology of PHAII. Furthermore, we identify renal distal bicarbonate secretion as a novel mechanism of renal tubular acidosis.


Asunto(s)
Acidosis Tubular Renal/fisiopatología , Túbulos Renales Colectores/fisiopatología , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/complicaciones , Transportadores de Sulfato/metabolismo , Acidosis Tubular Renal/sangre , Acidosis Tubular Renal/etiología , Animales , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Túbulos Renales Colectores/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación Missense , Potasio/sangre , Potasio/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Eliminación Renal , Cloruro de Sodio/metabolismo , Simportadores de Sodio-Bicarbonato/metabolismo , Transportadores de Sulfato/genética , Regulación hacia Arriba
6.
Nature ; 482(7383): 98-102, 2012 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-22266938

RESUMEN

Hypertension affects one billion people and is a principal reversible risk factor for cardiovascular disease. Pseudohypoaldosteronism type II (PHAII), a rare Mendelian syndrome featuring hypertension, hyperkalaemia and metabolic acidosis, has revealed previously unrecognized physiology orchestrating the balance between renal salt reabsorption and K(+) and H(+) excretion. Here we used exome sequencing to identify mutations in kelch-like 3 (KLHL3) or cullin 3 (CUL3) in PHAII patients from 41 unrelated families. KLHL3 mutations are either recessive or dominant, whereas CUL3 mutations are dominant and predominantly de novo. CUL3 and BTB-domain-containing kelch proteins such as KLHL3 are components of cullin-RING E3 ligase complexes that ubiquitinate substrates bound to kelch propeller domains. Dominant KLHL3 mutations are clustered in short segments within the kelch propeller and BTB domains implicated in substrate and cullin binding, respectively. Diverse CUL3 mutations all result in skipping of exon 9, producing an in-frame deletion. Because dominant KLHL3 and CUL3 mutations both phenocopy recessive loss-of-function KLHL3 mutations, they may abrogate ubiquitination of KLHL3 substrates. Disease features are reversed by thiazide diuretics, which inhibit the Na-Cl cotransporter in the distal nephron of the kidney; KLHL3 and CUL3 are expressed in this location, suggesting a mechanistic link between KLHL3 and CUL3 mutations, increased Na-Cl reabsorption, and disease pathogenesis. These findings demonstrate the utility of exome sequencing in disease gene identification despite the combined complexities of locus heterogeneity, mixed models of transmission and frequent de novo mutation, and establish a fundamental role for KLHL3 and CUL3 in blood pressure, K(+) and pH homeostasis.


Asunto(s)
Proteínas Portadoras/genética , Proteínas Cullin/genética , Hipertensión/genética , Mutación/genética , Seudohipoaldosteronismo/genética , Desequilibrio Hidroelectrolítico/genética , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Presión Sanguínea/genética , Proteínas Portadoras/química , Estudios de Cohortes , Proteínas Cullin/química , Electrólitos , Exones/genética , Femenino , Perfilación de la Expresión Génica , Genes Dominantes/genética , Genes Recesivos/genética , Genotipo , Homeostasis/genética , Humanos , Concentración de Iones de Hidrógeno , Hipertensión/complicaciones , Hipertensión/fisiopatología , Masculino , Ratones , Proteínas de Microfilamentos , Modelos Moleculares , Datos de Secuencia Molecular , Fenotipo , Potasio/metabolismo , Seudohipoaldosteronismo/complicaciones , Seudohipoaldosteronismo/fisiopatología , Cloruro de Sodio/metabolismo , Desequilibrio Hidroelectrolítico/complicaciones , Desequilibrio Hidroelectrolítico/fisiopatología
7.
Curr Opin Nephrol Hypertens ; 25(5): 417-23, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27322883

RESUMEN

PURPOSE OF REVIEW: Abundant evidence supports that the NaCl cotransporter (NCC) activity is tightly regulated by the with-no-lysine (WNK) kinases. Here, we summarize the data regarding NCC regulation by WNKs, with a particular emphasis on WNK4. RECENT FINDINGS: Several studies involving in-vivo and in-vitro models have provided paradoxical data regarding WNK4 regulation of the NCC. Although some studies show that WNK4 can activate the NCC, other equally compelling studies show that WNK4 inhibits the NCC. Recent studies have shown that WNK4 is regulated by the intracellular chloride concentration ([Cl]i), which could account for these paradoxical results. In conditions of high [Cl]i, WNK4 could act as an inhibitor via heterodimer formation with other WNKs. In contrast, when [Cl]i is low, WNK4 can activate Ste20-related, proline-alanine-rich kinase (SPAK)/oxidative stress responsive kinase 1 (OSR1) and thus the NCC. Modulation of WNK4 by [Cl]i has been shown to account for the potassium-sensing properties of the distal convoluted tubule. Other regulators of WNK4 include hormones and ubiquitination. SUMMARY: Modulation of WNK4 activity by [Cl]i can account for its dual role on the NCC, and this has important physiological implications regarding the regulation of extracellular potassium concentration. Defective regulation of WNKs by ubiquitination explains most cases of familial hyperkalemic hypertension.


Asunto(s)
Proteínas Serina-Treonina Quinasas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Cloruros/metabolismo , Humanos , Túbulos Renales Distales/metabolismo , Potasio/metabolismo , Seudohipoaldosteronismo/fisiopatología
8.
Pediatr Nephrol ; 30(11): 1903-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25503323

RESUMEN

Gordon Syndrome (GS) is a rare familial hypertension syndrome with a characteristic hyperkalaemia which distinguishes it from other syndromic forms of hypertension that typically cause hypokalaemia. Patients with GS respond to aggressive salt-restriction or relatively small doses of thiazide diuretics, which suggests that activation of the thiazide-sensitive Na/Cl cotransporter (NCC) in the distal nephron is to blame. However, the mechanism has proved to be complex. In 2001, mutations in genes encoding two serine/threonine kinases, WNK1 and WNK4, were identified as causing GS. However, it took several years to appreciate that these kinases operated in a cascade with downstream serine/threonine kinases (SPAK and OSR1) actually phosphorylating and activating NCC and the closely related cotransporters NKCC1 and NKCC2. The hyperkalaemia in GS arises from an independent action of WNK1/WNK4 to reduce cell-surface expression of ROMK, the secretory K-channel in the collecting ducts. However, mutations in WNK1/4 are present in a small minority of GS families, and further genes have emerged (CUL3 and KLHL3) that code for Cullin-3 (a scaffold protein in an ubiquitin-E3 ligase) and an adaptor protein, Kelch3, respectively. These new players regulate the ubiquitination and proteasomal degradation of WNK kinases, thereby adding to the complex picture we now have of NCC regulation in the distal nephron.


Asunto(s)
Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Humanos
9.
Curr Opin Nephrol Hypertens ; 23(5): 487-93, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24992566

RESUMEN

PURPOSE OF REVIEW: Efforts to explore the pathogenic mechanisms underlying hereditary hypertension caused by a single gene mutation have brought about conceptual advances in our understanding of blood pressure regulation. We here discuss a novel pathogenic mechanism underlying the hereditary hypertensive disease pseudohypoaldosteronism type II (PHAII), caused by mutations in three different genes encoding for Cullin-3, Kelch-like protein 3 (KLHL3), and with-no-lysine kinases (WNKs). RECENT FINDINGS: In 2001, mutations in genes encoding for WNKs were identified as being responsible for PHAII. Recent advancements in genetics, in particular whole-exome sequencing, have revealed that mutations in two additional genes encoding for KLHL3 and Cyllin3 also cause PHAII. This discovery contributed to the clarification of the previously unknown regulatory mechanism of WNKs, namely WNK ubiquitination by the KLHL3-Cullin-3 E3 ligase complex. SUMMARY: Levels of WNKs within cells are regulated via ubiquitination by the KLHL3-Cullin-3 E3 ligase complex and are important determinants of the activity of the WNK-oxidative stress-responsive gene 1 and Ste20-related proline-alanine-rich kinase-SLC12A transporter signaling cascade. The PHAII-causing mutations in WNK4, KLHL3, and Cullin-3 result in the decreased ubiquitination and increased abundance of WNK4 in the kidney, thereby activating the thiazide-sensitive NaCl cotransporter and causing PHAII.


Asunto(s)
Presión Sanguínea , Proteínas Cullin/metabolismo , Riñón/enzimología , Seudohipoaldosteronismo/enzimología , Equilibrio Hidroelectrolítico , Proteínas Adaptadoras Transductoras de Señales , Animales , Presión Sanguínea/genética , Proteínas Portadoras/metabolismo , Proteínas Cullin/genética , Predisposición Genética a la Enfermedad , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/fisiopatología , Proteínas de Microfilamentos , Antígenos de Histocompatibilidad Menor , Mutación , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Transducción de Señal , Proteína Quinasa Deficiente en Lisina WNK 1 , Equilibrio Hidroelectrolítico/genética
11.
Hum Mol Genet ; 20(5): 855-66, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21131289

RESUMEN

WNK1 (with-no-lysine[K]-1) is a protein kinase of which mutations cause a familial hypertension and hyperkalemia syndrome known as pseudohypoaldosteronism type 2 (PHA2). Kidney-specific (KS) WNK1 is an alternatively spliced form of WNK1 kinase missing most of the kinase domain. KS-WNK1 downregulates the Na(+)-Cl(-) cotransporter NCC by antagonizing the effect of full-length WNK1 when expressed in Xenopus oocytes. The physiological role of KS-WNK1 in the regulation of NCC and potentially other Na(+) transporters in vivo is unknown. Here, we report that mice overexpressing KS-WNK1 in the kidney exhibited renal Na(+) wasting, elevated plasma levels of angiotensin II and aldosterone yet lower blood pressure relative to wild-type littermates. Immunofluorescent staining revealed reduced surface expression of total and phosphorylated NCC and the Na(+)-K(+)-2Cl(-) cotransporter NKCC2 in the distal convoluted tubule and the thick ascending limb of Henle's loop, respectively. Conversely, mice with targeted deletion of exon 4A (the first exon for KS-WNK1) exhibited Na(+) retention, elevated blood pressure on a high-Na(+) diet and increased surface expression of total and phosphorylated NCC and NKCC2 in respective nephron segments. Thus, KS-WNK1 is a negative regulator of NCC and NKCC2 in vivo and plays an important role in the control of Na(+) homeostasis and blood pressure. These results have important implications to the pathogenesis of PHA2 with WNK1 mutations.


Asunto(s)
Regulación hacia Abajo , Silenciador del Gen , Riñón/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/metabolismo , Receptores de Droga/genética , Simportadores de Cloruro de Sodio-Potasio/genética , Simportadores/genética , Secuencia de Aminoácidos , Animales , Presión Sanguínea , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Humanos , Riñón/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , Especificidad de Órganos , Proteínas Serina-Treonina Quinasas/genética , Estructura Terciaria de Proteína , Seudohipoaldosteronismo/enzimología , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Receptores de Droga/metabolismo , Sodio/metabolismo , Simportadores de Cloruro de Sodio-Potasio/química , Simportadores de Cloruro de Sodio-Potasio/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/metabolismo , Proteína Quinasa Deficiente en Lisina WNK 1
12.
J Cell Sci ; 124(Pt 9): 1391-5, 2011 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-21486947

RESUMEN

We recently reported increased phosphorylation of the NaCl cotransporter (NCC) in Wnk4(D561A/+) knock-in mice, an ideal model of the human hereditary hypertensive disease pseudohypoaldosteronism type II (PHAII). Although previous in vitro studies had suggested the existence of a phosphorylation cascade involving the WNK, OSR1 and SPAK kinases, whether the WNK-OSR1/SPAK cascade is in fact fully responsible for NCC phosphorylation in vivo and whether the activation of this cascade is the sole mediator of PHAII remained to be determined. To clarify these issues, we mated the Wnk4(D561A/+) knock-in mice with Spak and Osr1 knock-in mice in which the T-loop threonine residues in SPAK and OSR1 (243 and 185, respectively) were mutated to alanine to prevent activation by WNK kinases. We found that NCC phosphorylation was almost completely abolished in Wnk4(D561A/+)Spak(T)(243A/T243A)Osr1(T185A/+) triple knock-in mice, clearly demonstrating that NCC phosphorylation in vivo is dependent on the WNK-OSR1/SPAK cascade. In addition, the high blood pressure, hyperkalemia and metabolic acidosis observed in Wnk4(D561A/+) mice were corrected in the triple knock-in mice. These results clearly establish that PHAII caused by the WNK4 D561A mutation is dependent on the activation of the WNK-OSR1/SPAK-NCC cascade and that the contribution of other mechanisms to PHAII (independent of the WNK-OSR1/SPAK cascade) could be minimal.


Asunto(s)
Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/metabolismo , Animales , Presión Sanguínea/genética , Presión Sanguínea/fisiología , Immunoblotting , Ratones , Mutación Missense/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/fisiopatología
13.
Clin Sci (Lond) ; 124(12): 701-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23336180

RESUMEN

Mutations in the novel serine/threonine WNK [With No lysine (=K)] kinases WNK1 and WNK4 cause PHAII (pseudohypoaldosteronism type II or Gordon's syndrome), a rare monogenic syndrome which causes hypertension and hyperkalaemia on a background of a normal glomerular filtration rate. Current animal models for PHAII recapitulate some aspects of the disease phenotype, but give no clues to how rapidly the phenotype emerges or whether it is reversible. To this end we have created an inducible PHAII transgenic animal model that expresses a human disease-causing WNK4 mutation, WNK4 Q565E, under the control of the Tet-On system. Several PHAII inducible transgenic mouse lines were created, each with differing TG (transgene) copy numbers and displaying varying degrees of TG expression (low, medium and high). Each of these transgenic lines demonstrated similar elevations of BP (blood pressure) and plasma potassium after 4 weeks of TG induction. Withdrawal of doxycycline switched off mutant TG expression and the disappearance of the PHAII phenotype. Western blotting of microdissected kidney nephron segments confirmed that expression of the thiazide-sensitive NCC (Na⁺-Cl⁻ co-transporter) was increased, as expected, in the distal convoluted tubule when transgenic mice were induced with doxycycline. The kidneys of these mice also do not show the morphological changes seen in the previous transgenic model expressing the same mutant form of WNK4. This inducible model shows, for the first time, that in vivo expression of a mutant WNK4 protein is sufficient to cause the rapid and reversible appearance of a PHAII disease phenotype in mice.


Asunto(s)
Riñón/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/enzimología , Animales , Biomarcadores/sangre , Presión Sanguínea , Western Blotting , Modelos Animales de Enfermedad , Doxiciclina/farmacología , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Genotipo , Humanos , Hibridación Fluorescente in Situ , Riñón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Fenotipo , Potasio/sangre , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/sangre , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Simportadores del Cloruro de Sodio/metabolismo , Factores de Tiempo
14.
Endocr J ; 60(3): 299-304, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23197115

RESUMEN

Pseudohypoaldosteronism type 1 (PHA1) is a rare condition characterized by neonatal salt loss with elevated plasma aldosterone and renin levels. Two types of PHA1 have been described: an autosomal recessive systemic form and an autosomal dominant renal form, in which the target organ defect is confined to the renal tubules. The dominant renal form of PHA1 is caused by heterozygous mutations in the NR3C2 gene, which encodes the mineralocorticoid receptor (MR). We determined clinical and biochemical parameters in two familial and four sporadic Japanese patient and analyzed the status of the NR3C2 gene. Failure to thrive was noted in five of the six patients. In one of the familial cases, the mother had an episode of failure to thrive when she was a toddler, but received no medical treatment. NaCl supplementation was discontinued in four of the six patients after they reached one year of age and they have grown normally thereafter. However, in one patient, 9 g/day of salt has been required to maintain serum Na concentration after 1 year of age. Analysis of NR3C2 identified three novel mutations [c. C1951T (p.R651X), c.304_305delGC (p.A102fsX103), c.del 603A (p.T201fsX34)] and one previously reported mutation [c.A2839G (p.947X)]. p.R651X was identified in one familial case and one unrelated sporadic patient. The patient who has been supplemented with large amount of salt was heterozygous for c.del 603A in exon 2. In conclusion, our study expands the spectrum of phenotypes, and characterized mutations of NR3C2 in the renal form of PHA1.


Asunto(s)
Túbulos Renales/fisiopatología , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Aldosterona/sangre , Insuficiencia de Crecimiento/genética , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Japón , Masculino , Mutación , Fenotipo , Seudohipoaldosteronismo/terapia , Receptores de Mineralocorticoides/genética , Renina/sangre , Cloruro de Sodio/administración & dosificación
15.
Cell Metab ; 5(5): 331-44, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17488636

RESUMEN

WNK1 and WNK4 mutations have been reported to cause pseudohypoaldosteronism type II (PHAII), an autosomal-dominant disorder characterized by hyperkalemia and hypertension. To elucidate the molecular pathophysiology of PHAII, we generated Wnk4(D561A/+) knockin mice presenting the phenotypes of PHAII. The knockin mice showed increased apical expression of phosphorylated Na-Cl cotransporter (NCC) in the distal convoluted tubules. Increased phosphorylation of the kinases OSR1 and SPAK was also observed in the knockin mice. Apical localization of the ROMK potassium channel and transepithelial Cl(-) permeability in the cortical collecting ducts were not affected in the knockin mice, whereas activity of epithelial Na(+) channels (ENaC) was increased. This increase, however, was not evident after hydrochlorothiazide treatment, suggesting that the regulation of ENaC was not a genetic but a secondary effect. Thus, the pathogenesis of PHAII caused by a missense mutation of WNK4 was identified to be increased function of NCC through activation of the OSR1/SPAK-NCC phosphorylation cascade.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/fisiopatología , Animales , Análisis Químico de la Sangre , Presión Sanguínea , Canales Epiteliales de Sodio/metabolismo , Vectores Genéticos/genética , Túbulos Renales Distales/metabolismo , Ratones , Microscopía Fluorescente , Mutación Missense/genética , Fosforilación , Canales de Potasio de Rectificación Interna/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Simportadores del Cloruro de Sodio/metabolismo , Orina/química
16.
Pflugers Arch ; 463(3): 477-85, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22200850

RESUMEN

WNK is a serine/threonine kinase. Mutation in WNK1 or WNK4 kinase results in pseudohypoaldosteronism type II (PHA II) featuring hypertension, hyperkalemia and metabolic acidosis. Sodium chloride cotransporter (NCC) is known to be regulated by phosphorylation and trafficking. Dietary salt and hormonal stimulation, such as aldosterone, also affect the regulation of NCC. We have previously reported that WNK4 inhibits NCC protein expression. To determine whether dietary salt affects NCC abundance through WNK4-mediated mechanism, we investigated the effects of dietary salt change with or without aldosterone infusion (1 mg/kg/day) on NCC and WNK4 expression in rats. We found that high-salt (HS, 4% NaCl) diet significantly inhibits NCC mRNA expression and protein abundance while enhancing WNK4 mRNA and protein expression, whereas low-salt (LS, 0.07% NaCl) diet increases NCC mRNA expression and protein abundance while reducing WNK4 expression. We also found that aldosterone infusion in HS-fed rats increases NCC mRNA expression and protein abundance, but decreases WNK4 expression. Administration with spironolactone (0.1 g/kg/day) in LS-fed rats decreases NCC mRNA expression and protein abundance while increasing WNK4 expression. We further showed that ERK1/2 phosphorylation was increased in HS-fed rats, but decreased in LS-fed rats. In HEK293 cells, over-expressed WNK4 increases ERK1/2 phosphorylation, whereas knockdown of WNK4 expression decreases ERK1/2 phosphorylation. Aldosterone treatment for 3 h decreases ERK1/2 phosphorylation. These data suggest that dietary salt change affects NCC protein abundance in an aldosterone-dependent mechanism likely via the WNK4-ERK1/2-mediated pathway.


Asunto(s)
Aldosterona/fisiología , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Seudohipoaldosteronismo/fisiopatología , Simportadores del Cloruro de Sodio/biosíntesis , Cloruro de Sodio Dietético/administración & dosificación , Aldosterona/farmacología , Animales , Células HEK293 , Humanos , ARN Mensajero/metabolismo , Ratas
17.
Histochem Cell Biol ; 137(3): 339-53, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22207244

RESUMEN

Epithelial sodium channels (ENaCs) are located on the apical surface of cells and funnel Na(+) ions from the lumen into the cell. ENaC function also regulates extracellular fluid volume as water flows across membranes accompanying Na(+) ions to maintain osmolarity. To examine the sites of expression and intracellular localization of ENaC, we generated polyclonal antibodies against the extracellular domain of human α-ENaC subunit that we expressed in E. coli. Three-dimensional (3D) confocal microscopy of immunofluorescence using these antibodies for the first time revealed that ENaCs are uniformly distributed on the ciliary surface in all epithelial cells with motile cilia lining the bronchus in human lung and female reproductive tract, all along the fimbrial end of the fallopian tube, the ampulla and rare cells in the uterine glands. Quantitative analysis indicated that cilia increase cell surface area >70-fold and the amount of ENaC on cilia is >1,000-fold higher than on non-ciliated cell surface. These findings indicate that ENaC functions as a regulator of the osmolarity of the periciliary fluid bathing the cilia. In contrast to ENaC, cystic fibrosis transmembrane conductance regulator (CFTR) that channels chloride ions from the cytoplasm to the lumen is located mainly on the apical side, but not on cilia. The cilial localization of ENaC requires reevaluation of the mechanisms of action of CFTR and other modulators of ENaC function. ENaC on motile cilia should be essential for diverse functions of motile cilia, such as germ cell transport, fertilization, implantation, clearance of respiratory airways and cell migration.


Asunto(s)
Cilios/fisiología , Canales Epiteliales de Sodio/genética , Canales Epiteliales de Sodio/metabolismo , Trompas Uterinas/fisiología , Mucosa Respiratoria/fisiología , Animales , Axonema/fisiología , Bronquios/fisiología , Bovinos , Línea Celular , Clonación Molecular , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Femenino , Expresión Génica/fisiología , Humanos , Ratones , Oviductos/fisiología , Seudohipoaldosteronismo/metabolismo , Seudohipoaldosteronismo/fisiopatología , Sodio/metabolismo , Spodoptera , Tubulina (Proteína)/metabolismo
18.
Clin Exp Nephrol ; 16(4): 530-8, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22294159

RESUMEN

BACKGROUND: We found that a mechanism of hypertension in pseudohypoaldosteronism type II (PHAII) caused by a WNK4 missense mutation (D561A) was activation of the WNK-OSR1/SPAK-NCC signal cascade. However, the pathogenic effect of intronic deletions in WNK1 genes also observed in PHAII patients remains unclear. To understand the pathophysiological roles of WNK1 in vivo, WNK1(+/-)mice have been analyzed, because homozygous WNK1 knockout is embryonic lethal. Although WNK1(+/-) mice have been reported to have hypotension, detailed analyses of the WNK signal cascade in the kidney and other organs of WNK1(+/-) mice have not been performed. METHOD: We assess the effect of heterozygous deletion of WNK1 on the WNK-OSR1/SPAK-NCC/NKCC1/NKCC2 signal cascade in the kidney and blood vessels. RESULTS: Contrary to the previous report, the blood pressure of WNK1(+/-) mice was not decreased, even under a low-salt diet. Under a WNK4(D561A/+) background, the heterozygous deletion of the WNK1 gene did not reduce the high blood pressure either. We then evaluated the phosphorylation status of OSR1, SPAK, NCC, NKCC1, and NKCC2 in the kidney, but no significant decrease in the phosphorylation was observed in WNK1(+/-) mice or WNK1(+/-)WNK4(D561A/+) mice. In contrast, a significant decrease in NKCC1 phosphorylation in the aorta and a decreased pressure-induced myogenic response in the mesenteric arteries were observed in WNK1(+/-) mice. CONCLUSION: The contribution of WNK1 to total WNK kinase activity in the kidney may be small, but that WNK1 may play a substantial role in the regulation of blood pressure in the arteries.


Asunto(s)
Vasos Sanguíneos/fisiología , Eliminación de Gen , Heterocigoto , Riñón/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Transducción de Señal/fisiología , Animales , Presión Sanguínea/fisiología , Modelos Animales de Enfermedad , Endotelio Vascular/fisiopatología , Hipertensión/fisiopatología , Riñón/irrigación sanguínea , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Músculo Liso Vascular/fisiopatología , Seudohipoaldosteronismo/fisiopatología , Receptores de Droga/fisiología , Simportadores de Cloruro de Sodio-Potasio/fisiología , Miembro 1 de la Familia de Transportadores de Soluto 12 , Miembro 2 de la Familia de Transportadores de Soluto 12 , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/fisiología , Proteína Quinasa Deficiente en Lisina WNK 1
19.
Pflugers Arch ; 462(6): 767-77, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21927811

RESUMEN

The thiazide-sensitive NaCl cotransporter (NCC) plays key roles in renal electrolyte transport and blood pressure maintenance. Regulation of this cotransporter has received increased attention recently, prompted by the discovery that mutations in the with-no-lysine (WNK) kinases are the molecular explanation for pseudohypoaldosteronism type II (PHAII). Studies suggest that WNK4 regulates NCC via two distinct pathways, depending on its state of activation. Furthermore, an intact STE20-related proline-alanine-rich kinase (SPAK)/oxidative stress response 1 kinase (OSR1) pathway was found to be necessary for a WNK4 PHAII mutation to increase NCC phosphorylation and blood pressure in mice. The mouse protein 25α is a novel regulator of the SPAK/OSR1 kinase family, which greatly increases their activity. The phosphorylation status of NCC and the WNK is regulated by the serum- and glucocorticoid-inducible kinase 1, suggesting novel mechanisms whereby aldosterone modulates NCC activity. Dephosphorylation of NCC by protein phosphatase 4 strongly influences the activity of the cotransporter, confirming an important role for NCC phosphorylation. Finally, γ-adducin increases NCC activity. This stimulatory effect is dependent on the phosphorylation status of the cotransporter. γ-Adducin only binds the dephosphorylated cotransporter, suggesting that phosphorylation of NCC causes the dissociation of γ-adducin. Since γ-adducin is not a kinase, it is tempting to speculate that the protein exerts its function by acting as a scaffold between the dephosphorylated cotransporter and the regulatory kinase. As more molecular regulators of NCC are identified, the system-controlling NCC activity is becoming increasingly complex. This intricacy confers an ability to integrate a variety of stimuli, thereby regulating NCC transport activity and ultimately blood pressure.


Asunto(s)
Riñón/metabolismo , Receptores de Droga/metabolismo , Cloruro de Sodio/metabolismo , Simportadores/metabolismo , Tiazidas/metabolismo , Animales , Presión Sanguínea/fisiología , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas de Unión a Calmodulina/genética , Proteínas de Unión a Calmodulina/metabolismo , Humanos , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Modelos Moleculares , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Fosfoproteínas Fosfatasas/genética , Fosfoproteínas Fosfatasas/metabolismo , Conformación Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/fisiopatología , Receptores de Droga/química , Receptores de Droga/genética , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/química , Simportadores/genética
20.
Hum Mol Genet ; 18(20): 3978-86, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19633012

RESUMEN

We recently generated Wnk4(D561A/+) knockin mice and found that a major pathogenesis of pseudohypoaldosteronism type II was the activation of the OSR1/SPAK kinase-NaCl cotransporter (NCC) phosphorylation cascade by the mutant WNK4. However, the physiological roles of wild-type WNK4 on the regulation of Na excretion and blood pressure, and whether wild-type WNK4 functions positively or negatively in this cascade, remained to be determined. In the present study, we generated WNK4 hypomorphic mice by deleting exon 7 of the Wnk4 gene. These mice did not show hypokalemia and metabolic alkalosis, but they did exhibit low blood pressure and increased Na and K excretion under low-salt diet. Phosphorylation of OSR1/SPAK and NCC was significantly reduced in the mutant mice as compared with their wild-type littermates. Protein levels of ROMK and Maxi K were not changed, but epithelial Na channel appeared to be activated as a compensatory mechanism for the reduced NCC function. Thus, wild-type WNK4 is a positive regulator for the WNK-OSR1/SPAK-NCC cascade, and WNK4 is a potential target of anti-hypertensive drugs.


Asunto(s)
Presión Sanguínea , Silenciador del Gen , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/metabolismo , Receptores de Droga/metabolismo , Sodio/metabolismo , Simportadores/metabolismo , Secuencia de Aminoácidos , Animales , Transporte Biológico , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fosforilación , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/fisiopatología , Receptores de Droga/genética , Miembro 3 de la Familia de Transportadores de Soluto 12 , Simportadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA