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










Base de datos
Intervalo de año de publicación
1.
Hypertension ; 81(4): 801-810, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38258567

RESUMEN

BACKGROUND: Potassium regulates the WNK (with no lysine kinase)-SPAK (STE20/SPS1-related proline/alanine-rich kinase) signaling axis, which in turn controls the phosphorylation and activation of the distal convoluted tubule thiazide-sensitive NCC (sodium-chloride cotransporter) for sodium-potassium balance. Although their roles in the kidney have not been investigated, it has been postulated that Cab39 (calcium-binding protein 39) or Cab39l (Cab39-like) is required for SPAK/OSR1 (oxidative stress response 1) activation. This study demonstrates how they control the WNK-SPAK/OSR1-NCC pathway. METHODS: We created a global knockout of Cab39l and a tamoxifen-inducible, NCC-driven, Cab39 knockout. The 2 lines were crossed to generate Cab39-DKO (Cab39 double knockout) animals. Mice were studied under control and low-potassium diet, which activates WNK-SPAK/OSR1-NCC phosphorylation. Western blots were used to assess the expression and phosphorylation of proteins. Blood and urine electrolytes were measured to test for compromised NCC function. Immunofluorescence studies were conducted to localize SPAK and OSR1. RESULTS: Both Cab39l and Cab39 are expressed in distal convoluted tubule, and only the elimination of both leads to a striking absence of NCC phosphorylation. Cab39-DKO mice exhibited a loss-of-NCC function, like in Gitelman syndrome. In contrast to the apical membrane colocalization of SPAK with NCC in wild-type mice, SPAK and OSR1 become confined to intracellular puncta in the Cab39-DKO mice. CONCLUSIONS: In the absence of Cab39 proteins, NCC cannot be phosphorylated, resulting in a Gitelman-like phenotype. Cab39 proteins function to localize SPAK at the apical membrane with NCC, reminiscent of the Cab39 yeast homolog function, translocating kinases during cytokinesis.


Asunto(s)
Proteínas Serina-Treonina Quinasas , Tiazidas , Ratones , Animales , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/genética , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Tiazidas/farmacología , Fosforilación , Túbulos Renales Distales/metabolismo , Potasio/metabolismo
2.
Curr Opin Nephrol Hypertens ; 32(5): 482-489, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37530088

RESUMEN

PURPOSE OF REVIEW: We recently localized a new K-Cl cotransporters-3 (KCC3) transporter to the apical membrane of type-B intercalated cells. This gives us an opportunity to revisit the roles of the KCC3 in kidney and integrate the new findings to our current knowledge of the biology of the bicarbonate secreting cells. RECENT FINDINGS: Here, we review the basic properties of the K-Cl cotransporter with a particular attention to the responsiveness of the transporter to cell swelling. We summarize what is already known about KCC3b and discuss new information gained from our localizing of KCC3a in type-B intercalated cells. We integrate the physiology of KCC3a with the main function of the type-B cell, that is, bicarbonate secretion through the well characterized apical Cl-/HCO3- exchanger and the basolateral Na-HCO3 cotransporter. SUMMARY: Both KCC3b and KCC3a seem to be needed for maintaining cell volume during enhanced inward cotransport of Na-glucose in proximal tubule and Na-HCO3 in intercalated cells. In addition, apical KCC3a might couple to pendrin function to recycle Cl-, particularly in conditions of low salt diet and therefore low Cl- delivery to the distal tubule. This function is critical in alkalemia, and KCC3a function in the pendrin-expressing cells may contribute to the K+ loss which is observed in alkalemia.


Asunto(s)
Bicarbonatos , Simportadores , Animales , Humanos , Bicarbonatos/metabolismo , Riñón/metabolismo , Proteínas de Transporte de Membrana , Transportadores de Sulfato , Mamíferos/metabolismo , Cotransportadores de K Cl
3.
Am J Physiol Renal Physiol ; 325(3): F377-F393, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37498547

RESUMEN

The urinary potassium (K+) excretion machinery is upregulated with increasing dietary K+, but the role of accompanying dietary anions remains inadequately characterized. Poorly absorbable anions, including [Formula: see text], are thought to increase K+ secretion through a transepithelial voltage effect. Here, we tested if they also influence the K+ secretion machinery. Wild-type mice, aldosterone synthase (AS) knockout (KO) mice, or pendrin KO mice were randomized to control, high-KCl, or high-KHCO3 diets. The K+ secretory capacity was assessed in balance experiments. Protein abundance, modification, and localization of K+-secretory transporters were evaluated by Western blot analysis and confocal microscopy. Feeding the high-KHCO3 diet increased urinary K+ excretion and the transtubular K+ gradient significantly more than the high-KCl diet, coincident with more pronounced upregulation of epithelial Na+ channels (ENaC) and renal outer medullary K+ (ROMK) channels and apical localization in the distal nephron. Experiments in AS KO mice revealed that the enhanced effects of [Formula: see text] were aldosterone independent. The high-KHCO3 diet also uniquely increased the large-conductance Ca2+-activated K+ (BK) channel ß4-subunit, stabilizing BKα on the apical membrane, the Cl-/[Formula: see text] exchanger, pendrin, and the apical KCl cotransporter (KCC3a), all of which are expressed specifically in pendrin-positive intercalated cells. Experiments in pendrin KO mice revealed that pendrin was required to increase K+ excretion with the high-KHCO3 diet. In summary, [Formula: see text] stimulates K+ excretion beyond a poorly absorbable anion effect, upregulating ENaC and ROMK in principal cells and BK, pendrin, and KCC3a in pendrin-positive intercalated cells. The adaptive mechanism prevents hyperkalemia and alkalosis with the consumption of alkaline ash-rich diets but may drive K+ wasting and hypokalemia in alkalosis.NEW & NOTEWORTHY Dietary anions profoundly impact K+ homeostasis. Here, we found that a K+-rich diet, containing [Formula: see text] as the counteranion, enhances the electrogenic K+ excretory machinery, epithelial Na+ channels, and renal outer medullary K+ channels, much more than a high-KCl diet. It also uniquely induces KCC3a and pendrin, in B-intercalated cells, providing an electroneutral KHCO3 secretion pathway. These findings reveal new K+ balance mechanisms that drive adaption to alkaline and K+-rich foods, which should guide new treatment strategies for K+ disorders.


Asunto(s)
Alcalosis , Potasio , Animales , Ratones , Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Aniones/metabolismo , Dieta , Ratones Noqueados , Potasio/metabolismo , Potasio en la Dieta/metabolismo , Sodio/metabolismo , Transportadores de Sulfato/genética
4.
Am J Physiol Cell Physiol ; 324(5): C1171-C1178, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-37036298

RESUMEN

A primary function of intercalated cells in the distal tubule of the kidney is to maintain pH homeostasis. For example, type B intercalated cells secrete bicarbonate largely through the action of the apical Cl-/HCO3- exchanger, pendrin, which helps correct metabolic alkalosis. Since both the K-Cl cotransporter, KCC3a and pendrin colocalize to the apical region of type B and non-A, non-B intercalated cells and since both are upregulated in models of metabolic alkalosis, such as with dietary NaHCO3 loading, we raised the possibility that apical KCC3a facilitates pendrin-mediated bicarbonate secretion, such as through apical Cl- recycling. The purpose of this study was to determine if KCC3a abundance changes through intake of bicarbonate alone or through bicarbonate plus its accompanying cation, and if it requires a direct interaction with pendrin or the renin-angiotensin-aldosterone system. We observed that KCC3a protein abundance, but not mRNA, increases in a mouse model of metabolic alkalosis, achieved with dietary NaHCO3 or KHCO3 intake. Bicarbonate ion increases KCC3a abundance, both in vivo and in vitro, independently of the accompanying cation. Moreover, bicarbonate intake upregulates KCC3a independently of aldosterone or angiotensin II. Since NaHCO3 intake increased KCC3a abundance in wild-type as well as in pendrin knockout mice, this KCC3a upregulation by bicarbonate does not depend on a direct interaction with pendrin. We conclude that increased extracellular bicarbonate, as observed in models of metabolic alkalosis, directly raises KCC3a abundance independently of angiotensin II, aldosterone, or changes in KCC3a transcription and does not involve a direct interaction with pendrin.NEW & NOTEWORTHY KCC3a expression is stimulated in alkalemia. This paper shows that bicarbonate itself is mediating this effect through a posttranscriptional mechanism. The paper also shows that this phenomenon is not mediated by aldosterone or angiotensin II.


Asunto(s)
Alcalosis , Bicarbonatos , Animales , Ratones , Bicarbonatos/metabolismo , Aldosterona/farmacología , Aldosterona/metabolismo , Angiotensina II/farmacología , Angiotensina II/metabolismo , Riñón/metabolismo , Transportadores de Sulfato/genética , Transportadores de Sulfato/metabolismo , Alcalosis/metabolismo , Proteínas de Transporte de Anión/genética
5.
J Am Soc Nephrol ; 33(3): 584-600, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35064051

RESUMEN

BACKGROUND: Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) gene cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) increases abundance of With-No-Lysine (K) Kinase 4 (WNK4), inappropriately activating sterile 20/SPS-1-related proline/alanine-rich kinase (SPAK), which then phosphorylates and hyperactivates the Na+Cl- cotransporter (NCC). The precise mechanism by which CUL3-Δ9 causes FHHt is unclear. We tested the hypothesis that reduced abundance of CUL3 and of Kelch-like 3 (KLHL3), the CUL3 substrate adaptor for WNK4, is mechanistically important. Because JAB1, an enzyme that inhibits CUL3 activity by removing the ubiquitin-like protein NEDD8, cannot interact with CUL3-Δ9, we also determined whether Jab1 disruption mimicked the effects of CUL3-Δ9 expression. METHODS: We used an inducible renal tubule-specific system to generate several mouse models expressing CUL3-Δ9, mice heterozygous for both CUL3 and KLHL3 (Cul3+/-/Klhl3+/- ), and mice with short-term Jab1 disruption (to avoid renal injury associated with long-term disruption). RESULTS: Renal KLHL3 was higher in Cul3-/- mice, but lower in Cul3-/-/Δ9 mice and in the Cul3+/-/Δ9 FHHt model, suggesting KLHL3 is a target for both WT and mutant CUL3. Cul3+/-/Klhl3+/- mice displayed increased WNK4-SPAK activation and phospho-NCC abundance and an FHHt-like phenotype with increased plasma [K+] and salt-sensitive blood pressure. Short-term Jab1 disruption in mice lowered the abundance of CUL3 and KLHL3 and increased the abundance of WNK4 and phospho-NCC. CONCLUSIONS: Jab1-/- mice and Cul3+/-/Klhl3+/- mice recapitulated the effects of CUL3-Δ9 expression on WNK4-SPAK-NCC. Our data suggest degradation of both KLHL3 and CUL3 plays a central mechanistic role in CUL3-Δ9-mediated FHHt.


Asunto(s)
Proteínas Cullin , Hipertensión , Seudohipoaldosteronismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Femenino , Humanos , Hipertensión/genética , Masculino , Ratones , Proteínas de Microfilamentos/genética , Proteínas Serina-Treonina Quinasas/genética , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
6.
Cells ; 11(1)2021 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-35011657

RESUMEN

The thiazide-sensitive sodium chloride cotransporter (NCC) plays a vital role in maintaining sodium (Na+) and potassium (K+) homeostasis. NCC activity is modulated by with-no-lysine kinases 1 and 4 (WNK1 and WNK4), the abundance of which is controlled by the RING-type E3 ligase Cullin 3 (Cul3) and its substrate adapter Kelch-like protein 3. Dietary K+ intake has an inverse correlation with NCC activity, but the mechanism underlying this phenomenon remains to be fully elucidated. Here, we investigated the involvement of other members of the cullin family in mediating K+ effects on NCC phosphorylation (active form) and abundance. In kidneys from mice fed diets varying in K+ content, there were negative correlations between NCC (phosphorylated and total) and active (neddylated) forms of cullins (Cul1, 3, 4, and 5). High dietary K+ effects on phosphorylated NCC were attenuated in Cul3 mutant mice (CUL3-Het/Δ9). Short-term (30 min) and long-term (24 h) alterations in the extracellular K+ concentration did not affect cullin neddylation levels in ex vivo renal tubules. In the short term, the ability of high extracellular K+ to decrease NCC phosphorylation was preserved in the presence of MLN4924 (pan-cullin inhibitor), but the response to low extracellular K+ was absent. In the long term, MLN4924 attenuated the effects of high extracellular K+ on NCC phosphorylation, and responses to low extracellular K+ were absent. Our data suggest that in addition to Cul3, other cullins are involved in mediating the effects of K+ on NCC phosphorylation and abundance.


Asunto(s)
Proteínas Cullin/metabolismo , Potasio/farmacología , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Ciclopentanos/farmacología , Suplementos Dietéticos , Túbulos Renales/efectos de los fármacos , Túbulos Renales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Fosforilación/efectos de los fármacos , Pirimidinas/farmacología
7.
Hypertension ; 76(5): 1461-1469, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32981364

RESUMEN

Increased sympathoexcitation and renal sodium retention during high salt intake are hallmarks of the salt sensitivity of blood pressure. The mechanism(s) by which excessive sympathetic nervous system release of norepinephrine influences renal sodium reabsorption is unclear. However, studies demonstrate that norepinephrine can stimulate the activity of the NCC (sodium chloride cotransporter) and promote the development of SSH (salt-sensitive hypertension). The adrenergic signaling pathways governing NCC activity remain a significant source of controversy with opposing studies suggesting a central role of upstream α1- and ß-adrenoceptors in the canonical regulatory pathway involving WNKs (with-no-lysine kinases), SPAK (STE20/SPS1-related proline alanine-rich kinase), and OxSR1 (oxidative stress response 1). In our previous study, α1-adrenoceptor antagonism in norepinephrine-infused male Sprague-Dawley rats prevented the development of norepinephrine-evoked SSH in part by suppressing NCC activity and expression. In these studies, we used selective adrenoceptor antagonism in male Dahl salt-sensitive rats to test the hypothesis that norepinephrine-mediated activation of the NCC in Dahl SSH occurs via an α1-adrenoceptor dependent pathway. A high-salt diet evoked significant increases in NCC activity, expression, and phosphorylation in Dahl salt-sensitive rats that developed SSH. Increases were associated with a dysfunctional WNK1/4 dynamic and a failure to suppress SPAK/OxSR1 activity. α1-adrenoceptor antagonism initiated before high-salt intake or following the establishment of SSH attenuated blood pressure in part by suppressing NCC activity, expression, and phosphorylation. Collectively, our findings support the existence of a norepinephrine-activated α1-adrenoceptor gated pathway that relies on WNK/SPAK/OxSR1 signaling to regulate NCC activity in SSH.


Asunto(s)
Regulación de la Expresión Génica , Hipertensión/metabolismo , Simportadores del Cloruro de Sodio/metabolismo , Sistema Nervioso Simpático/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacología , Antagonistas Adrenérgicos beta/farmacología , Animales , Presión Sanguínea/efectos de los fármacos , Presión Sanguínea/fisiología , Hipertensión/genética , Hipertensión/fisiopatología , Masculino , Fosforilación/efectos de los fármacos , Prazosina/análogos & derivados , Prazosina/farmacología , Propranolol/farmacología , Ratas , Ratas Endogámicas Dahl , Ratas Sprague-Dawley , Simportadores del Cloruro de Sodio/genética , Sistema Nervioso Simpático/efectos de los fármacos , Sistema Nervioso Simpático/fisiopatología
8.
Kidney Int ; 97(1): 95-105, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31623859

RESUMEN

Acute cardiorenal syndrome is a common complication of acute cardiovascular disease. Studies of acute kidney injury (AKI) to chronic kidney disease (CKD) transition, including patients suffering acute cardiovascular disease, report high rates of CKD development. Therefore, acute cardiorenal syndrome associates with CKD, but no study has established causation. To define this we used a murine cardiac arrest (CA) and cardiopulmonary resuscitation (CPR) model or sham procedure on male mice. CA was induced with potassium chloride while CPR consisted of chest compressions and epinephrine eight minutes later. Two weeks after AKI was induced by CA/CPR, the measured glomerular filtration rate (GFR) was not different from sham. However, after seven weeks the mice developed CKD, recapitulating clinical observations. One day, and one, two, and seven weeks after CA/CPR, the GFR was measured, and renal tissue sections were evaluated for various indices of injury and inflammation. One day after CA/CPR, acute cardiorenal syndrome was indicated by a significant reduction of the mean GFR (649 in sham, vs. 25 µL/min/100g in CA/CPR animals), KIM-1 positive tubules, and acute tubular necrosis. Renal inflammation developed, with F4/80 positive and CD3-positive cells infiltrating the kidney one day and one week after CA/CPR, respectively. Although there was functional recovery with normalization of GFR two weeks after CA/CPR, deposition of tubulointerstitial matrix proteins α-smooth muscle actin and fibrillin-1 progressed, along with a significantly reduced mean GFR (623 in sham vs. 409 µL/min/100g in CA/CPR animals), proteinuria, increased tissue transforming growth factor-ß, and fibrosis establishing the development of CKD seven weeks after CA/CPR. Thus, murine CA/CPR, a model of acute cardiorenal syndrome, causes an AKI-CKD transition likely due to prolonged renal inflammation.


Asunto(s)
Lesión Renal Aguda/inmunología , Síndrome Cardiorrenal/inmunología , Túbulos Renales/patología , Nefritis/inmunología , Insuficiencia Renal Crónica/inmunología , Lesión Renal Aguda/patología , Animales , Síndrome Cardiorrenal/patología , Reanimación Cardiopulmonar , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Fibrosis , Tasa de Filtración Glomerular/inmunología , Paro Cardíaco/inducido químicamente , Paro Cardíaco/complicaciones , Paro Cardíaco/inmunología , Paro Cardíaco/terapia , Humanos , Inflamación/inmunología , Inflamación/patología , Túbulos Renales/inmunología , Masculino , Ratones , Nefritis/patología , Cloruro de Potasio/administración & dosificación , Cloruro de Potasio/toxicidad , Insuficiencia Renal Crónica/patología
9.
Physiol Rep ; 7(17): e14195, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31496133

RESUMEN

The distal nephron is essential for calcium homeostasis. This is evidenced by disordered calcium transport following disrupted distal nephron function occurring in salt-wasting tubulopathies or with diuretic use. A plethora of studies support a role for WNK4 in thick ascending limb (TAL) and distal convoluted tubule ion transport with most studies focusing on sodium transport. Little is known about the in vivo role of WNK4 in regulating calcium homeostsis. Here, we investigated the role of WNK4 in regulating distal nephron calcium transport using WNK4 knockout animals (WNK4-/- ). As has been shown previously, we found that baseline urinary calcium levels are normal following WNK4 deletion. Following acute treatment with the loop diuretic, furosemide, which causes hypercalciuria through TAL inhibition, WNK4-/- animals demonstrated increased calcium wasting compared with wild-type controls. WNK4-/- animals had decreased TRPV5 expression along DCT2 supporting a mechanistic role for this calcium channel in the increased calciuresis. As this supported the hypothesis that WNK4-/- animals have a tendency toward calcium wasting under stress, we tested the effects of a calcium-deplete diet on urinary calcium excretion. Urinary calcium excretion and plasma ionized calcium levels were not different between control and knockout animals following consumption of a calcium-deplete diet. Our data show that WNK4, via regulation of TRPV5, limits distal calcium losses following acute treatment with furosemide; however, WNK4 deletion does not affect the chronic renal response to dietary calcium depletion. Our data reveal an in vivo role for WNK4 in distal nephron calcium handling that is important for fine-tuning calcium reabsorption.


Asunto(s)
Calcio de la Dieta/orina , Túbulos Renales Proximales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Insuficiencia Renal/metabolismo , Animales , Canales de Calcio/genética , Canales de Calcio/metabolismo , Calcio de la Dieta/metabolismo , Diuréticos/toxicidad , Furosemida/toxicidad , Túbulos Renales Proximales/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/genética , Eliminación Renal , Insuficiencia Renal/etiología , Sodio/metabolismo , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo
10.
Am J Physiol Renal Physiol ; 317(4): F825-F838, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31364380

RESUMEN

Hypomagnesemia is associated with reduced kidney function and life-threatening complications and sustains hypokalemia. The distal convoluted tubule (DCT) determines final urinary Mg2+ excretion and, via activity of the Na+-Cl- cotransporter (NCC), also plays a key role in K+ homeostasis by metering Na+ delivery to distal segments. Little is known about the mechanisms by which plasma Mg2+ concentration regulates NCC activity and how low-plasma Mg2+ concentration and K+ concentration interact to modulate NCC activity. To address this, we performed dietary manipulation studies in mice. Compared with normal diet, abundances of total NCC and phosphorylated NCC (pNCC) were lower after short-term (3 days) or long-term (14 days) dietary Mg2+ restriction. Altered NCC activation is unlikely to play a role, since we also observed lower total NCC abundance in mice lacking the two NCC-activating kinases, STE20/SPS-1-related proline/alanine-rich kinase and oxidative stress response kinase-1, after Mg2+ restriction. The E3 ubiquitin-protein ligase NEDD4-2 regulates NCC abundance during dietary NaCl loading or K+ restriction. Mg2+ restriction did not lower total NCC abundance in inducible nephron-specific neuronal precursor cell developmentally downregulated 4-2 (NEDD4-2) knockout mice. Total NCC and pNCC abundances were similar after short-term Mg2+ or combined Mg2+-K+ restriction but were dramatically lower compared with a low-K+ diet. Therefore, sustained NCC downregulation may serve a mechanism that enhances distal Na+ delivery during states of hypomagnesemia, maintaining hypokalemia. Similar results were obtained with long-term Mg2+-K+ restriction, but, surprisingly, NCC was not activated after long-term K+ restriction despite lower plasma K+ concentration, suggesting significant differences in distal tubule adaptation to acute or chronic K+ restriction.


Asunto(s)
Hipopotasemia/metabolismo , Deficiencia de Magnesio/metabolismo , Ubiquitina-Proteína Ligasas Nedd4/biosíntesis , Animales , Dieta , Regulación hacia Abajo , Túbulos Renales Distales/metabolismo , Magnesio/sangre , Deficiencia de Magnesio/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ubiquitina-Proteína Ligasas Nedd4/genética , Fosforilación , Potasio/sangre , Deficiencia de Potasio/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/biosíntesis , Miembro 3 de la Familia de Transportadores de Soluto 12/genética
11.
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
12.
Sci Rep ; 9(1): 4596, 2019 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-30872636

RESUMEN

Cullin 3 (CUL3) is part of the ubiquitin proteasomal system and controls several cellular processes critical for normal organ function including the cell cycle, and Keap1/Nrf2 signaling. Kidney tubule-specific Cul3 disruption causes tubulointerstitial fibrosis, but little is known about the mechanisms. Therefore, we tested the hypothesis that dysregulation of the cell cycle and Keap1/Nrf2 pathway play a role in initiating the kidney injury upon Cul3 disruption. Cul3 deletion increased expression of cyclin E and p21, associated with uncontrolled proliferation, DNA damage, and apoptosis, all of which preceded proximal tubule injury. The cdk2-cyclin E inhibitor roscovitine did not prevent the effects of Cul3 deletion, but instead exacerbated the kidney injury. Injury occurred despite accumulation and activation of CUL3 substrate Keap1/Nrf2, proposed to be protective in kidney injury. Cul3 disruption led to progressive interstitial inflammation, functionally relevant renal fibrosis and death. Finally, we observed reduced CUL3 expression in several AKI and CKD mouse models and in fibrotic human kidney tissue. These data establish CUL3 knockout mice as a novel genetic CKD model in which dysregulation of the cell cycle may play a primary role in initiating tubule injury, and that CUL3 dysregulation could contribute to acute and fibrotic kidney disease.


Asunto(s)
Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Eliminación de Gen , Enfermedades Renales/genética , Enfermedades Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Túbulos Renales Proximales/patología , Animales , Biomarcadores , Línea Celular , Proliferación Celular , Daño del ADN , Modelos Animales de Enfermedad , Fibrosis , Técnica del Anticuerpo Fluorescente , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Inmunohistoquímica , Proteína 1 Asociada A ECH Tipo Kelch/metabolismo , Enfermedades Renales/mortalidad , Enfermedades Renales/patología , Ratones , Ratones Noqueados , Factor 2 Relacionado con NF-E2/metabolismo , Insuficiencia Renal/genética , Insuficiencia Renal/metabolismo , Insuficiencia Renal/mortalidad , Insuficiencia Renal/patología , Transducción de Señal , Ubiquitinación
13.
Am J Physiol Renal Physiol ; 315(4): F781-F790, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29412704

RESUMEN

With no lysine kinase 4 (WNK4) is essential to activate the thiazide-sensitive NaCl cotransporter (NCC) along the distal convoluted tubule, an effect central to the phenotype of familial hyperkalemic hypertension. Although effects on potassium and sodium channels along the connecting and collecting tubules have also been documented, WNK4 is typically believed to have little role in modulating sodium chloride reabsorption along the thick ascending limb of the loop of Henle. Yet wnk4-/- mice (knockout mice lacking WNK4) do not demonstrate the hypocalciuria typical of pure distal convoluted tubule dysfunction. Here, we tested the hypothesis that WNK4 also modulates bumetanide-sensitive Na-K-2Cl cotransporter (NKCC2) function along the thick ascending limb. We confirmed that w nk4-/- mice are hypokalemic and waste sodium chloride, but are also normocalciuric. Results from Western blots suggested that the phosphorylated forms of both NCC and NKCC2 were in lower abundance in wnk4-/- mice than in controls. This finding was confirmed by immunofluorescence microscopy. Although the initial response to furosemide was similar in wnk4-/- mice and controls, the response was lower in the knockout mice when reabsorption along the distal convoluted tubule was inhibited. Using HEK293 cells, we showed that WNK4 increases the abundance of phosphorylated NKCC2. More supporting evidence that WNK4 may modulate NKCC2 emerges from a mouse model of WNK4-mediated familial hyperkalemic hypertension in which more phosphorylated NKCC2 is present than in controls. These data indicate that WNK4, in addition to modulating NCC, also modulates NKCC2, contributing to its physiological function in vivo.


Asunto(s)
Cloruros/metabolismo , Túbulos Renales Distales/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Animales , Hipertensión/metabolismo , Túbulos Renales Colectores/metabolismo , Lisina/metabolismo , Ratones Noqueados , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Simportadores de Cloruro de Sodio-Potasio/metabolismo
14.
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
15.
Physiol Genomics ; 50(2): 104-116, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29212850

RESUMEN

Hypertension poses a significant challenge to vasculature homeostasis and stands as the most common cardiovascular disease in the world. Its effects are especially profound on endothelial cells that form the inner lining of the vasculature and are directly exposed to the effects of excess pressure. Here, we characterize the in vivo transcriptomic response of cardiac endothelial cells to hypertension by rapidly isolating these cells from the spontaneous hypertension mouse model BPH/2J and its normotensive BPN/3J control strain and performing and RNA sequencing on both. Comparison of transcriptional differences between these groups reveals statistically significant changes in cellular pathways consistent with cardiac fibrosis found in hypertensive animals. Importantly, many of the fibrosis-linked genes identified also differ significantly between juvenile prehypertensive and adult hypertensive BPH/2J mice, suggesting that these transcriptional differences are hypertension related. We examined the dynamic nature of these transcriptional changes by testing whether blood pressure normalization using either a calcium channel blocker (amlodipine) or a angiotensin II receptor blocker (losartan) is able to reverse these expression patterns associated with hypertension. We find that blood pressure reduction is capable of reversing some gene-expression patterns, while other transcripts are recalcitrant to therapeutic intervention. This illuminates the possibility that unmanaged hypertension may irreversibly alter some endothelial transcriptional patterns despite later intervention. This study quantifies how endothelial cells are remodeled at the molecular level in cardiovascular pathology and advances our understanding of the transcriptional events associated with endothelial response to hypertensive challenge.


Asunto(s)
Fibrosis/metabolismo , Frecuencia Cardíaca/efectos de los fármacos , Hipertensión/metabolismo , Amlodipino/uso terapéutico , Animales , Presión Sanguínea/efectos de los fármacos , Bloqueadores de los Canales de Calcio/uso terapéutico , Modelos Animales de Enfermedad , Fibrosis/genética , Frecuencia Cardíaca/genética , Hipertensión/tratamiento farmacológico , Hipertensión/genética , Losartán/uso terapéutico , Masculino , Ratones
16.
JCI Insight ; 2(24)2017 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-29263298

RESUMEN

Mutations in the ubiquitin ligase scaffold protein Cullin 3 (CUL3) cause the disease familial hyperkalemic hypertension (FHHt). In the kidney, mutant CUL3 (CUL3-Δ9) increases abundance of With-No-Lysine [K] Kinase 4 (WNK4), with excessive activation of the downstream Sterile 20 (STE20)/SPS-1-related proline/alanine-rich kinase (SPAK) increasing phosphorylation of the Na+-Cl- cotransporter (NCC). CUL3-Δ9 promotes its own degradation via autoubiquitination, leading to the hypothesis that Cul3 haploinsufficiency causes FHHt. To directly test this, we generated Cul3 heterozygous mice (CUL3-Het), and Cul3 heterozygotes also expressing CUL3-Δ9 (CUL3-Het/Δ9), using an inducible renal epithelial-specific system. Endogenous CUL3 was reduced to 50% in both models, and consistent with autoubiquitination, CUL3-Δ9 protein was undetectable in CUL3-Het/Δ9 kidneys unless primary renal epithelia cells were cultured. Abundances of WNK4 and phosphorylated NCC did not differ between control and CUL3-Het mice, but they were elevated in CUL3-Het/Δ9 mice, which also displayed higher plasma [K+] and blood pressure. Abundance of phosphorylated Na+-K+-2Cl- cotransporter (NKCC2) was also increased, which may contribute to the severity of CUL3-Δ9-mediated FHHt. WNK4 and SPAK localized to puncta in NCC-positive segments but not in NKCC2-positive segments, suggesting differential effects of CUL3-Δ9. These results indicate that Cul3 haploinsufficiency does not cause FHHt, but dominant effects of CUL3-Δ9 are required.


Asunto(s)
Proteínas Cullin/genética , Proteínas Cullin/metabolismo , Seudohipoaldosteronismo/genética , Seudohipoaldosteronismo/metabolismo , Animales , Presión Sanguínea/genética , Células Cultivadas , Células Epiteliales , Femenino , Haploinsuficiencia , Heterocigoto , Riñón/metabolismo , Masculino , Ratones , Mutación , Fosforilación , Potasio/sangre , Proteínas Serina-Treonina Quinasas/metabolismo , Seudohipoaldosteronismo/fisiopatología , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo , Ubiquitinación , Proteína Wnt4/metabolismo
17.
Am J Physiol Renal Physiol ; 310(11): F1389-96, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27076645

RESUMEN

Chronic high blood pressure (hypertension) is the most common disease in the Unites States. While several classes of drugs exist to treat it, many patients (up to 10 million Americans) respond poorly to therapy, even when multiple classes are used. Recent evidence suggests that a significant portion of patients will always remain hypertensive despite maximum therapy with the drugs currently available. Therefore, there is a pressing need to develop novel antihypertensive agents. One limitation has been the identification of new targets, a limitation that has been overcome by recent insights into the mechanisms underlying monogenic forms of hypertension. The disease familial hyperkalemic hypertension is caused by mutations in with-no-lysine (WNK) kinases 1 and 4 and in cullin-3 and kelch-like 3, components of an E3 ubiquitin ligase complex that promotes WNK kinase degradation. The study of the mechanisms by which this pathway regulates blood pressure has identified several candidates for the development of new antihypertensive agents. This pathway is particularly attractive since its inhibition may not only reduce renal sodium reabsorption along multiple segments but may also reduce vascular tone. Here, we will describe the mechanisms by which this pathway regulate blood pressure and discuss the potential of targeting it to develop new antihypertensive drugs.


Asunto(s)
Antihipertensivos/uso terapéutico , Proteínas Portadoras/metabolismo , Proteínas Cullin/metabolismo , Hipertensión/tratamiento farmacológico , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Humanos , Hipertensión/metabolismo , Proteínas de Microfilamentos
18.
J Physiol ; 594(17): 4945-66, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27068441

RESUMEN

KEY POINTS: STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) phosphorylate and activate the renal Na(+) -K(+) -2Cl(-) cotransporter 2 (NKCC2) and Na(+) Cl(-) cotransporter (NCC). Mouse models suggest that OSR1 mainly activates NKCC2-mediated sodium transport along the thick ascending limb, while SPAK mainly activates NCC along the distal convoluted tubule, but the kinases may compensate for each other. We hypothesized that disruption of both kinases would lead to polyuria and severe salt-wasting, and generated SPAK/OSR1 double knockout mice to test this. Despite a lack of SPAK and OSR1, phosphorylated NKCC2 abundance was still high, suggesting the existence of an alternative activating kinase. Compensatory changes in SPAK/OSR1-independent phosphorylation sites on both NKCC2 and NCC and changes in sodium transport along the collecting duct were also observed. Potassium restriction revealed that SPAK and OSR1 play essential roles in the emerging model that NCC activation is central to sensing changes in plasma [K(+) ]. ABSTRACT: STE20 (Sterile 20)/SPS-1 related proline/alanine-rich kinase (SPAK) and oxidative stress-response kinase-1 (OSR1) activate the renal cation cotransporters Na(+) -K(+) -2Cl(-) cotransporter (NKCC2) and Na(+) -Cl(-) cotransporter (NCC) via phosphorylation. Knockout mouse models suggest that OSR1 mainly activates NKCC2, while SPAK mainly activates NCC, with possible cross-compensation. We tested the hypothesis that disrupting both kinases causes severe polyuria and salt-wasting by generating SPAK/OSR1 double knockout (DKO) mice. DKO mice displayed lower systolic blood pressure compared with SPAK knockout (SPAK-KO) mice, but displayed no severe phenotype even after dietary salt restriction. Phosphorylation of NKCC2 at SPAK/OSR1-dependent sites was lower than in SPAK-KO mice, but still significantly greater than in wild type mice. In the renal medulla, there was significant phosphorylation of NKCC2 at SPAK/OSR1-dependent sites despite a complete absence of SPAK and OSR1, suggesting the existence of an alternative activating kinase. The distal convoluted tubule has been proposed to sense plasma [K(+) ], with NCC activation serving as the primary effector pathway that modulates K(+) secretion, by metering sodium delivery to the collecting duct. Abundance of phosphorylated NCC (pNCC) is dramatically lower in SPAK-KO mice than in wild type mice, and the additional disruption of OSR1 further reduced pNCC. SPAK-KO and kidney-specific OSR1 single knockout mice maintained plasma [K(+) ] following dietary potassium restriction, but DKO mice developed severe hypokalaemia. Unlike mice lacking SPAK or OSR1 alone, DKO mice displayed an inability to phosphorylate NCC under these conditions. These data suggest that SPAK and OSR1 are essential components of the effector pathway that maintains plasma [K(+) ].


Asunto(s)
Túbulos Renales Distales/metabolismo , Potasio/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Presión Sanguínea , Homeostasis , Túbulos Renales Distales/fisiología , Masculino , Ratones , Ratones Noqueados , Potasio/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Miembro 1 de la Familia de Transportadores de Soluto 12/metabolismo , Miembro 3 de la Familia de Transportadores de Soluto 12/metabolismo
19.
J Am Soc Nephrol ; 27(8): 2436-45, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26712527

RESUMEN

Excess aldosterone is an important contributor to hypertension and cardiovascular disease. Conversely, low circulating aldosterone causes salt wasting and hypotension. Aldosterone activates mineralocorticoid receptors (MRs) to increase epithelial sodium channel (ENaC) activity. However, aldosterone may also stimulate the thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC). Here, we generated mice in which MRs could be deleted along the nephron to test this hypothesis. These kidney-specific MR-knockout mice exhibited salt wasting, low BP, and hyperkalemia. Notably, we found evidence of deficient apical orientation and cleavage of ENaC, despite the salt wasting. Although these mice also exhibited deficient NCC activity, NCC could be stimulated by restricting dietary potassium, which also returned BP to control levels. Together, these results indicate that MRs regulate ENaC directly, but modulation of NCC is mediated by secondary changes in plasma potassium concentration. Electrolyte balance and BP seem to be determined, therefore, by a delicate interplay between direct and indirect mineralocorticoid actions in the distal nephron.


Asunto(s)
Túbulos Renales Distales/metabolismo , Receptores de Mineralocorticoides/fisiología , Cloruro de Sodio Dietético/metabolismo , Animales , Transporte Biológico , Ratones , Ratones Noqueados
20.
Exp Anim ; 61(2): 109-17, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22531725

RESUMEN

Preferences for different housing conditions in mice were evaluated by radiotelemetry. Male C57BL/6J and ICR mice were used. Preference for bedding materials in mice was compared among three materials, wood shavings (WS), paper (CF) and cloth (AG), using the length of stay in cages as a parameter. The results indicated that mice stayed longer in a cage with AG than in cages with other bedding materials. The present study confirmed our previous results and thereby indicated that radiotelemetry is a useful method to evaluate impacts of housing conditions on animal welfare. In the second part of this study, we used radiotelemetry to evaluate color preference of the mice for cloth bedding material. In C57BL/6J mice, staying time in black cloth was significantly longer than that in white cloth. In ICR mice, staying time in white cloth was significantly longer than that in black cloth. The mice preferred the environment with the same color as their fur, which may be important for animal welfare.


Asunto(s)
Crianza de Animales Domésticos , Animales de Laboratorio/psicología , Conducta Animal/fisiología , Conducta de Elección/fisiología , Vivienda para Animales , Telemetría/métodos , Bienestar del Animal , Animales , Color , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...