Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Opin Nephrol Hypertens ; 28(5): 474-480, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31313674

RESUMO

PURPOSE OF REVIEW: The apical Na/K/2Cl cotransporter (NKCC2) mediates NaCl reabsorption by the thick ascending limb, contributing to maintenance of blood pressure (BP). Despite effective NKCC2 inhibition by loop diuretics, these agents are not viable for long-term management of BP due to side effects. Novel molecular mechanisms that control NKCC2 activity reveal an increasingly complex picture with interacting layers of NKCC2 regulation. Here, we review the latest developments that shine new light on NKCC2-mediated control of BP and potential new long-term therapies to treat hypertension. RECENT FINDINGS: Emerging molecular NKCC2 regulators, often binding partners, reveal a complex overlay of interacting mechanisms aimed at fine tuning NKCC2 activity. Different factors achieve this by shifting the balance between trafficking steps like exocytosis, endocytosis, recycling and protein turnover, or by balancing phosphorylation vs. dephosphorylation. Further molecular details are also emerging on previously known pathways of NKCC2 regulation, and recent in-vivo data continues to place NKCC2 regulation at the center of BP control. SUMMARY: Several layers of emerging molecular mechanisms that control NKCC2 activity may operate simultaneously, but they can also be controlled independently. This provides an opportunity to identify new pharmacological targets to fine-tune NKCC2 activity for BP management.


Assuntos
Pressão Sanguínea/fisiologia , Hipertensão/tratamento farmacológico , Membro 1 da Família 12 de Carreador de Soluto/fisiologia , Animais , Antígenos de Neoplasias/fisiologia , Proteínas de Ciclo Celular/fisiologia , Humanos , Proteínas de Neoplasias/fisiologia , Fosforilação , Proteínas Serina-Treonina Quinases/fisiologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/uso terapêutico , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores
2.
Am J Physiol Renal Physiol ; 316(4): F743-F757, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30623725

RESUMO

Chronic adenine feeding is extensively used to develop animal models of chronic renal failure with metabolic features resembling those observed in humans. However, the mechanism by which adenine induces renal failure is poorly understood. In this study, we examined the early effects of adenine on water metabolism and salt balance in rats placed in metabolic cages and fed control or adenine-containing diets for 7 days. Molecular and functional studies demonstrated that adenine-fed rats exhibited a significant reduction in food intake, polyuria, polydipsia, decreased urine osmolality, and increased salt wasting. These effects are independent of changes in food intake and result from a coordinated downregulation of water channel aquaporin-2 (AQP2) and salt transporter (Na+-K+-Cl- cotransporter 2; NKCC2) in the collecting duct and medullary thick ascending limb, respectively. As a result, adenine-fed rats exhibited massive volume depletion, as indicated by a significant body weight loss, increased blood urea nitrogen, and increased hematocrit and hemoglobin levels, all of which were significantly corrected with NaCl replacement. Adenine-induced urinary concentrating defect was not corrected by exogenous arginine vasopressin (AVP), and it correlated with reduced cAMP production in vivo and in vitro. In conclusion, adenine acts on renal tubules as a signaling molecule and causes nephrogenic diabetes insipidus with salt wasting, at least, by directly interfering with AVP V2 receptor signaling with subsequent downregulation of NKCC2 and AQP2 in the kidney. The combination of renal fluid loss and decreased food intake with subsequent massive volume depletion likely plays an important role in the development of early prerenal failure that progresses to chronic kidney disease in long-term adenine feeding.


Assuntos
Adenina/toxicidade , Nefropatias/induzido quimicamente , Nefropatias/metabolismo , Rim/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Animais , Aquaporina 2/antagonistas & inibidores , Arginina Vasopressina/farmacologia , AMP Cíclico/metabolismo , Dieta , Relação Dose-Resposta a Droga , Ingestão de Alimentos , Rim/patologia , Nefropatias/patologia , Masculino , Concentração Osmolar , Ratos , Ratos Sprague-Dawley , Cloreto de Sódio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Água/metabolismo , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
3.
Kidney Int ; 94(4): 701-715, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30007527

RESUMO

Uromodulin, the most abundant protein in normal urine, is essentially produced by the cells lining the thick ascending limb. There it regulates the activity of the cotransporter NKCC2 and is involved in sodium chloride handling and blood pressure regulation. Conflicting reports suggested that uromodulin may also be expressed in the distal convoluted tubule (DCT) where its role remains unknown. Using microdissection studies combined with fluorescent in situ hybridization and co-immunostaining analyses, we found a significant expression of uromodulin in mouse and human DCT at approximately 10% of thick ascending limb expression levels, but restricted to the early part of the DCT (DCT1). Genetic deletion of Umod in mouse was reflected by a major shift in NCC activity from the DCT1 to the downstream DCT2 segment, paralleled by a compensatory expansion of DCT2. By increasing the distal sodium chloride and calcium ion load with chronic furosemide administration, an intrinsic compensatory defect in the DCT from Umod-/- compared to wild type mice was found manifested as sodium wasting and hypercalciuria. In line, co-expression studies in HEK cells suggested a facilitating role for uromodulin in NCC phosphorylation, possibly via SPAK-OSR1 modulation. These experiments demonstrate a significant expression of uromodulin in the early part of mouse and human DCT. Thus, biosynthesis of uromodulin in the DCT1 is critical for its function, structure and plasticity, suggesting novel links between uromodulin, blood pressure control and risk of kidney stones.


Assuntos
Túbulos Renais Distais/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Uromodulina/biossíntese , Uromodulina/genética , Uromodulina/metabolismo , Animais , Furosemida/farmacologia , Expressão Gênica , Células HEK293 , Humanos , Hipercalciúria/induzido quimicamente , Hipercalciúria/genética , Túbulos Renais Distais/fisiologia , Masculino , Camundongos , Camundongos Knockout , Fosforilação , RNA Mensageiro , Sódio/metabolismo , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/genética
4.
J Am Soc Nephrol ; 29(7): 1838-1848, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29848507

RESUMO

Background Hypercalciuria can result from activation of the basolateral calcium-sensing receptor (CaSR), which in the thick ascending limb of Henle's loop controls Ca2+ excretion and NaCl reabsorption in response to extracellular Ca2+ However, the function of CaSR in the regulation of NaCl reabsorption in the distal convoluted tubule (DCT) is unknown. We hypothesized that CaSR in this location is involved in activating the thiazide-sensitive NaCl cotransporter (NCC) to prevent NaCl loss.Methods We used a combination of in vitro and in vivo models to examine the effects of CaSR on NCC activity. Because the KLHL3-WNK4-SPAK pathway is involved in regulating NaCl reabsorption in the DCT, we assessed the involvement of this pathway as well.Results Thiazide-sensitive 22Na+ uptake assays in Xenopus laevis oocytes revealed that NCC activity increased in a WNK4-dependent manner upon activation of CaSR with Gd3+ In HEK293 cells, treatment with the calcimimetic R-568 stimulated SPAK phosphorylation only in the presence of WNK4. The WNK4 inhibitor WNK463 also prevented this effect. Furthermore, CaSR activation in HEK293 cells led to phosphorylation of KLHL3 and WNK4 and increased WNK4 abundance and activity. Finally, acute oral administration of R-568 in mice led to the phosphorylation of NCC.Conclusions Activation of CaSR can increase NCC activity via the WNK4-SPAK pathway. It is possible that activation of CaSR by Ca2+ in the apical membrane of the DCT increases NaCl reabsorption by NCC, with the consequent, well known decrease of Ca2+ reabsorption, further promoting hypercalciuria.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sódio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Ativação Enzimática/genética , Células HEK293 , Humanos , Imidazóis/farmacologia , Masculino , Camundongos , Proteínas dos Microfilamentos , Oócitos , Fenetilaminas/farmacologia , Fosforilação/efeitos dos fármacos , Propilaminas/farmacologia , Proteína Quinase C/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/genética , Pirrolidinas/farmacologia , Receptores de Detecção de Cálcio/genética , Transdução de Sinais , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Transfecção , Proteínas de Xenopus/metabolismo , Xenopus laevis
5.
Am J Physiol Renal Physiol ; 314(2): F240-F250, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29046296

RESUMO

Geranium seemannii Peyr is a perennial plant endemic to central Mexico that has been widely used for its diuretic effect, but the responsible compound of this effect is unknown as well as the mechanism by which the diuretic effect is achieved. Geraniin is one of the compounds isolated from this kind of geranium. This study was designed to determinate whether geraniin possesses diuretic activity and to elucidate the mechanism of action. Geraniin was extracted and purified from Geranium seemannii Peyr. Male Wistar rats were divided into four groups: 1) Control, 2) 75 mg/kg of geraniin, 3) 20 mg/kg of furosemide, and 4) 10 mg/kg of hydrochlorothiazide. Each treatment was administered by gavage every 24 h for 7 days. The urinary excretion of electrolytes and the fractional excretion of sodium (FENa) were determined. To uncover the molecular target of geraniin, Xenopus laevis oocytes were microinjected with cRNAs encoding the Na+-Cl- cotransporter (NCC) and the Na+-K+-2Cl- cotransporter NKCC2 to functionally express these cotransporters. Geraniin significantly increased diuresis, natriuresis, and calciuresis to a similar extent as was observed in the furosemide-treated rats. Consistent with the furosemide-like effect, in X. laevis oocytes, geraniin significantly reduced the activity of NKCC2, with no effect on NCC activity. In contrast to furosemide, the effect of geraniin on NKCC2 was irreversible, apparently due to its inhibitory effect on heat shock protein 90. Our observations suggest that geraniin could have a potential role in the treatment of hypertension or edematous states.


Assuntos
Diurese/efeitos dos fármacos , Diuréticos/farmacologia , Glucosídeos/farmacologia , Taninos Hidrolisáveis/farmacologia , Rim/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Animais , Biomarcadores/urina , Cálcio/urina , Relação Dose-Resposta a Droga , Furosemida/farmacologia , Proteínas de Choque Térmico HSP90/metabolismo , Rim/metabolismo , Masculino , Natriurese/efeitos dos fármacos , Ratos Wistar , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo , Xenopus laevis
6.
Am J Physiol Renal Physiol ; 313(6): F1209-F1215, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28835421

RESUMO

Afferent arteriole (Af-Art) resistance is modulated by two intrinsic nephron feedbacks: 1) the vasoconstrictor tubuloglomerular feedback (TGF) mediated by Na+-K+-2Cl- cotransporters (NKCC2) in the macula densa and blocked by furosemide and 2) the vasodilator connecting tubule glomerular feedback (CTGF), mediated by epithelial Na+ channels (ENaC) in the connecting tubule and blocked by benzamil. High salt intake reduces Af-Art vasoconstrictor ability in Dahl salt-sensitive rats (Dahl SS). Previously, we measured CTGF indirectly, by differences between TGF responses with and without CTGF inhibition. We recently developed a new method to measure CTGF more directly by simultaneously inhibiting NKCC2 and the Na+/H+ exchanger (NHE). We hypothesize that in vivo during simultaneous inhibition of NKCC2 and NHE, CTGF causes an Af-Art dilatation revealed by an increase in stop-flow pressure (PSF) in Dahl SS and that is enhanced with a high salt intake. In the presence of furosemide alone, increasing nephron perfusion did not change the PSF in either Dahl salt-resistant rats (Dahl SR) or Dahl SS. When furosemide and an NHE inhibitor, dimethylamiloride, were perfused simultaneously, an increase in tubular flow caused Af-Art dilatation that was demonstrated by an increase in PSF. This increase was greater in Dahl SS [4.5 ± 0.4 (SE) mmHg] than in Dahl SR (2.5 ± 0.3 mmHg; P < 0.01). We confirmed that CTGF causes this vasodilation, since benzamil completely blocked this effect. However, a high salt intake did not augment the Af-Art dilatation. We conclude that during simultaneous inhibition of NKCC2 and NHE in the nephron, CTGF induces Af-Art dilatation and a high salt intake failed to enhance this effect.


Assuntos
Arteríolas/fisiopatologia , Glomérulos Renais/irrigação sanguínea , Túbulos Renais/fisiopatologia , Circulação Renal , Cloreto de Sódio na Dieta/efeitos adversos , Vasodilatação , Amilorida/análogos & derivados , Amilorida/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/metabolismo , Canais Epiteliais de Sódio/efeitos dos fármacos , Canais Epiteliais de Sódio/metabolismo , Retroalimentação Fisiológica , Furosemida/farmacologia , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/metabolismo , Masculino , Ratos Endogâmicos Dahl , Circulação Renal/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Trocadores de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo , Resistência Vascular , Vasoconstrição , Vasodilatação/efeitos dos fármacos
7.
Am J Physiol Renal Physiol ; 312(6): F1120-F1127, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28228405

RESUMO

The Na+-K+-2Cl- cotransporter (NKCC2) on the loop of Henle is the site of action of furosemide. Because outer medullary potassium channel (ROMK) inhibitors prevent reabsorption by NKCC2, we tested the hypothesis that ROMK inhibition with a novel selective ROMK inhibitor (compound C) blocks tubuloglomerular feedback (TGF) and reduces vascular resistance. Loop perfusion of either ROMK inhibitor or furosemide caused dose-dependent blunting of TGF, but the response to furosemide was 10-fold more sensitive (IC50 = 10-6 M for furosemide and IC50 = 10-5 M for compound C). During systemic infusion, both diuretics inhibited TGF, but ROMK inhibitor was 10-fold more sensitive (compound C: 63% inhibition; furosemide: 32% inhibition). Despite blockade of TGF, 1 h of constant systemic infusion of both diuretics reduced the glomerular filtration rate (GFR) and renal blood flow (RBF) by 40-60% and increased renal vascular resistance (RVR) by 100-200%. Neither diuretic altered blood pressure or hematocrit. Proximal tubule hydrostatic pressures (PPT) increased transiently with both diuretics (compound C: 56% increase; furosemide: 70% increase) but returned to baseline. ROMK inhibitor caused more natriuresis (3,400 vs. 1,600% increase) and calciuresis (1,200 vs. 800% increase) but less kaliuresis (33 vs. 167% increase) than furosemide. In conclusion, blockade of ROMK or Na+-K+-2Cl- transport inhibits TGF yet increases renal vascular resistance. The renal vasoconstriction was independent of volume depletion, blood pressure, TGF, or PPT.


Assuntos
Diuréticos/farmacologia , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/irrigação sanguínea , Túbulos Renais/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/antagonistas & inibidores , Vasoconstrição/efeitos dos fármacos , Vasoconstritores/farmacologia , Anestesia Geral , Animais , Cálcio/urina , Relação Dose-Resposta a Droga , Retroalimentação , Furosemida/farmacologia , Taxa de Filtração Glomerular/efeitos dos fármacos , Pressão Hidrostática , Glomérulos Renais/metabolismo , Túbulos Renais/metabolismo , Masculino , Modelos Animais , Natriurese/efeitos dos fármacos , Potássio/urina , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Resistência Vascular/efeitos dos fármacos
8.
Am J Physiol Renal Physiol ; 311(6): F1217-F1229, 2016 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-27707706

RESUMO

Sodium and its associated anions are the major determinant of extracellular fluid volume, and the reabsorption of Na+ by the kidney plays a crucial role in long-term blood pressure control. The goal of this study was to investigate the extent to which inhibitors of transepithelial Na+ transport (TNa) along the nephron alter urinary solute excretion and TNa efficiency and how those effects may vary along different nephron segments. To accomplish that goal, we used the multinephron model developed in the companion study (28). That model represents detailed transcellular and paracellular transport processes along the nephrons of a rat kidney. We simulated the inhibition of the Na+/H+ exchanger (NHE3), the bumetanide-sensitive Na+-K+-2Cl- transporter (NKCC2), the Na+-Cl- cotransporter (NCC), and the amiloride-sensitive Na+ channel (ENaC). Under baseline conditions, NHE3, NKCC2, NCC, and ENaC reabsorb 36, 22, 4, and 7%, respectively, of filtered Na+ The model predicted that inhibition of NHE3 substantially reduced proximal tubule TNa and oxygen consumption (QO2 ). Whole-kidney TNa efficiency, as reflected by the number of moles of Na+ reabsorbed per moles of O2 consumed (denoted by the ratio TNa/QO2 ), decreased by ∼20% with 80% inhibition of NHE3. NKCC2 inhibition simulations predicted a substantial reduction in thick ascending limb TNa and QO2 ; however, the effect on whole-kidney TNa/QO2 was minor. Tubular K+ transport was also substantially impaired, resulting in elevated urinary K+ excretion. The most notable effect of NCC inhibition was to increase the excretion of Na+, K+, and Cl-; its impact on whole-kidney TNa and its efficiency was minor. Inhibition of ENaC was predicted to have opposite effects on the excretion of Na+ (increased) and K+ (decreased) and to have only a minor impact on whole-kidney TNa and TNa/QO2 Overall, model predictions agree well with measured changes in Na+ and K+ excretion in response to diuretics and Na+ transporter mutations.


Assuntos
Bloqueadores do Canal de Sódio Epitelial/farmacologia , Modelos Biológicos , Néfrons/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Animais , Transporte de Íons/efeitos dos fármacos , Néfrons/metabolismo , Ratos , Trocador 3 de Sódio-Hidrogênio
9.
Oncotarget ; 7(33): 53571-53582, 2016 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-27447551

RESUMO

A central aim in cancer research is to identify genes with altered expression patterns in tumor specimens and their potential role in tumorigenesis. Most types of tumors, including hepatocellular carcinoma (HCC), are heterogeneous in terms of genotype and phenotype. Thus, traditional analytical methods like the t-test fail to identify all oncogenes from expression profiles. In this study, we performed a meta-Cancer Outlier Profile Analysis (meta-COPA) across six microarray datasets for HCC from the GEO database. We found that gene SLC12A1 was overexpressed in the Hep3B cell line, compared with five other HCC cell lines and L02 cells. We also found that the upregulation of SLC12A1 was mediated by histone methylation within its promoter region, and that SLC12A1 is a positive regulator of the WNK1/ERK5 pathway. Consistent with in vitro results, treatment with the SLC12A1 antagonist Bumetanide delayed tumor formation and reduced Hep3B cell tumor size in mouse xenografts. In summary, our research reveals a novel subset of HCCs that are sensitive to SLC12A1 antagonist treatment, thereby offering a new strategy for precision HCC treatment.


Assuntos
Bumetanida/farmacologia , Carcinoma Hepatocelular/patologia , Neoplasias Hepáticas/patologia , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Animais , Bases de Dados Genéticas , Humanos , Camundongos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Am J Physiol Renal Physiol ; 310(10): F958-71, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26911852

RESUMO

Furosemide is a widely used, potent natriuretic drug, which inhibits the Na(+)-K(+)-2Cl(-) cotransporter (NKCC)-2 in the ascending limb of the loop of Henle applied to reduce extracellular fluid volume expansion in heart and kidney disease. Undesirable consequences of furosemide, such as worsening of kidney function and unpredictable effects on sodium balance, led to this critical evaluation of how inhibition of NKCC affects renal and cardiovascular physiology. This evaluation reveals important knowledge gaps, involving furosemide as a drug, the function of NKCC2 (and NKCC1), and renal and systemic indirect effects of NKCC inhibition. Regarding renal effects, renal blood flow and glomerular filtration rate could become compromised by activation of tubuloglomerular feedback or by renin release, particularly if renal function is already compromised. Modulation of the intrarenal renin angiotensin system, however, is ill-defined. Regarding systemic effects, vasodilation followed by nonspecific NKCC inhibition and changes in venous compliance are not well understood. Repetitive administration of furosemide induces short-term (braking phenomenon, acute diuretic resistance) and long-term (chronic diuretic resistance) adaptations, of which the mechanisms are not well known. Modulation of NKCC2 expression and activity in kidney and heart failure is ill-defined. Lastly, furosemide's effects on cutaneous sodium stores and on uric acid levels could be beneficial or detrimental. Concluding, a considerable knowledge gap is identified regarding a potent drug with a relatively specific renal target, NKCC2, and renal and systemic actions. Resolving these questions would increase the understanding of NKCCs and their actions and improve rational use of furosemide in pathophysiology of fluid volume expansion.


Assuntos
Furosemida/farmacologia , Natriurese/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Animais , Pressão Sanguínea/efeitos dos fármacos , Inibidores da Anidrase Carbônica/farmacologia , Resistência a Medicamentos , Antagonistas de Receptores de GABA-A/farmacologia , Humanos , Insuficiência Renal Crônica , Reabsorção Renal/efeitos dos fármacos , Sistema Renina-Angiotensina/efeitos dos fármacos
11.
J Biol Chem ; 291(9): 4487-502, 2016 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-26721884

RESUMO

Mutations in the renal specific Na-K-2Cl co-transporter (NKCC2) lead to type I Bartter syndrome, a life-threatening kidney disease featuring arterial hypotension along with electrolyte abnormalities. We have previously shown that NKCC2 and its disease-causing mutants are subject to regulation by endoplasmic reticulum-associated degradation (ERAD). The aim of the present study was to identify the protein partners specifically involved in ERAD of NKCC2. To this end, we screened a kidney cDNA library through a yeast two-hybrid assay using NKCC2 C terminus as bait. We identified OS9 (amplified in osteosarcomas) as a novel and specific binding partner of NKCC2. Co-immunoprecipitation assays in renal cells revealed that OS9 association involves mainly the immature form of NKCC2. Accordingly, immunocytochemistry analysis showed that NKCC2 and OS9 co-localize at the endoplasmic reticulum. In cells overexpressing OS9, total cellular NKCC2 protein levels were markedly decreased, an effect blocked by the proteasome inhibitor MG132. Pulse-chase and cycloheximide-chase assays demonstrated that the marked reduction in the co-transporter protein levels was essentially due to increased protein degradation of the immature form of NKCC2. Conversely, knockdown of OS9 by small interfering RNA increased NKCC2 expression by increasing the co-transporter stability. Inactivation of the mannose 6-phosphate receptor homology domain of OS9 had no effect on its action on NKCC2. In contrast, mutations of NKCC2 N-glycosylation sites abolished the effects of OS9, indicating that OS9-induced protein degradation is N-glycan-dependent. In summary, our results demonstrate the presence of an OS9-mediated ERAD pathway in renal cells that degrades immature NKCC2 proteins. The identification and selective modulation of ERAD components specific to NKCC2 and its disease-causing mutants might provide novel therapeutic strategies for the treatment of type I Bartter syndrome.


Assuntos
Degradação Associada com o Retículo Endoplasmático , Rim/metabolismo , Lectinas/metabolismo , Proteínas de Neoplasias/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Substituição de Aminoácidos , Animais , Síndrome de Bartter/genética , Síndrome de Bartter/metabolismo , Linhagem Celular , Degradação Associada com o Retículo Endoplasmático/efeitos dos fármacos , Biblioteca Gênica , Glicosilação/efeitos dos fármacos , Células HEK293 , Humanos , Imunoprecipitação , Rim/efeitos dos fármacos , Lectinas/antagonistas & inibidores , Lectinas/química , Lectinas/genética , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Gambás , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Inibidores de Proteassoma/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estabilidade Proteica/efeitos dos fármacos , Estrutura Terciária de Proteína , Interferência de RNA , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/química , Membro 1 da Família 12 de Carreador de Soluto/genética
12.
Ross Fiziol Zh Im I M Sechenova ; 101(4): 441-50, 2015 Apr.
Artigo em Russo | MEDLINE | ID: mdl-26336742

RESUMO

The hydrogen sulfide (H2S) influence on the contractile activity of vascular smooth muscle cells (SMC) was studied on endothelium-denuded aortic ring segments of male Wistar rats with method of mechanography. Contractions of SMS were induced by incubation in high potassium solution as well as in hyper-, hypo- and isosmotic solutions. 5-100 LM of H2S donor--sodium hydrosulfide (NaHS) increased mechanical tension of SMC precontracted with high potassium solution that was abolished by bumetanide--the inhibitor of Na+, K+, 2Cl(-) -cotransporter (NKCC), but 100-1000 microM of NaHS relaxed SMS. NaHS (10 microM) increased the amplitude of hyper- and isosmotic contraction, but not of hyposmotic contraction. NaHS (ImM) decreased the amplitude of hyper-, iso-, and hyposmotic contractions. The direct measurements of NKCC activity with radionuclide method showed an increase in NKCC activity under the action of 5-100 microM of NaHS. These findings suggest that low concentrations of H2S participate in the NKCC activation. This mechanism underlines constrictive action of H2S on smooth muscle cells.


Assuntos
Aorta Torácica/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Contração Muscular/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Vasoconstrição/efeitos dos fármacos , Animais , Aorta Torácica/citologia , Aorta Torácica/metabolismo , Aorta Torácica/fisiologia , Bumetanida/farmacologia , Tamanho Celular/efeitos dos fármacos , Soluções Hipertônicas , Soluções Hipotônicas , Técnicas In Vitro , Tono Muscular/efeitos dos fármacos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/fisiologia , Ratos Wistar , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores
13.
Sci Rep ; 5: 12480, 2015 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-26207612

RESUMO

Ureteral obstruction with subsequent hydronephrosis is a common clinical complication. Downregulation of renal sodium transporters in obstructed kidneys could contribute to impaired urinary concentrating capability and salt waste following the release of a ureteral obstruction. The current study was undertaken to investigate the role of mitochondrial complex-1 inhibition in modulating sodium transporters in obstructive kidney disease. Following unilateral ureteral obstruction (UUO) for 7 days, a global reduction of sodium transporters, including NHE3, α-Na-K-ATPase, NCC, NKCC2, p-NKCC2, ENaCα, and ENaCγ, was observed, as determined via qRT-PCR and/or Western blotting. Interestingly, inhibition of mitochondrial complex-1 by rotenone markedly reversed the downregulation of NKCC2, p-NKCC2, and ENaCα. In contrast, other sodium transporters were not affected by rotenone. To study the potential mechanisms involved in mediating the effects of rotenone on sodium transporters, we examined a number of known sodium modulators, including PGE2, ET1, Ang II, natriuretic peptides (ANP, BNP, and CNP), and nitric oxide synthases (iNOS, nNOS, and eNOS). Importantly, among these modulators, only BNP and iNOS were significantly reduced by rotenone treatment. Collectively, these findings demonstrated a substantial role of mitochondrial dysfunction in mediating the downregulation of NKCC2 and ENaCα in obstructive kidney disease, possibly via iNOS-derived nitric oxide and BNP.


Assuntos
Complexo I de Transporte de Elétrons/antagonistas & inibidores , Canais Epiteliais de Sódio/genética , Nefropatias/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Rotenona/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/genética , Angiotensina II/metabolismo , Animais , Dinoprostona/metabolismo , Complexo I de Transporte de Elétrons/genética , Complexo I de Transporte de Elétrons/metabolismo , Canais Epiteliais de Sódio/metabolismo , Regulação da Expressão Gênica , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Nefropatias/genética , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Peptídeo Natriurético Encefálico/metabolismo , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Óxido Nítrico Sintase Tipo II/metabolismo , Transdução de Sinais , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/genética , Trocadores de Sódio-Hidrogênio/metabolismo , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/agonistas , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo , Obstrução Ureteral/genética , Obstrução Ureteral/metabolismo , Obstrução Ureteral/patologia
14.
FEBS J ; 282(19): 3786-98, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26183401

RESUMO

Sodium chloride reabsorption in the thick ascending limb of the loop of Henle is mediated by the Na(+)-K(+)-2Cl(-) cotransporter (NKCC2). The loop diuretic furosemide is a potent inhibitor of NKCC2. However, less is known about the mechanism regulating the electrolyte transporter. Considering the well-established effects of nitric oxide on NKCC2 activity, cGMP is likely involved in this regulation. cGMP-dependent protein kinase I (cGKI; PKGI) is a cGMP target protein that phosphorylates different substrates after activation through cGMP. We investigated the potential correlation between the cGMP/cGKI pathway and NKCC2 regulation. We treated wild-type (wt) and cGKIα-rescue mice with furosemide. cGKIα-rescue mice expressed cGKIα only under the control of the smooth muscle-specific transgelin (SM22) promoter in a cGKI deficient background. Furosemide treatment increased the urine excretion of sodium and chloride in cGKIα-rescue mice compared to that in wt mice. We analyzed the phosphorylation of NKCC2 by western blotting and immunostaining using the phosphospecific antibody R5. The administration of furosemide significantly increased the phosphorylated NKCC2 signal in wt but not in cGKIα-rescue mice. NKCC2 activation led to its phosphorylation and membrane translocation. To examine whether cGKI was involved in this process, we analyzed vasodilator-stimulated phosphoprotein, which is phosphorylated by cGKI. Furosemide injection resulted in increased vasodilator-stimulated phosphoprotein phosphorylation in wt mice. We hypothesize that furosemide administration activated cGKI, leading to NKCC2 phosphorylation and membrane translocation. This cGKI-mediated pathway could be a mechanism to compensate for the inhibitory effect of furosemide on NKCC2.


Assuntos
Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Furosemida/farmacologia , Rim/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , GMP Cíclico/metabolismo , GMP Cíclico/urina , Proteína Quinase Dependente de GMP Cíclico Tipo I/genética , Feminino , Rim/efeitos dos fármacos , Masculino , Camundongos Knockout , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Fosfoproteínas/metabolismo , Fosforilação/efeitos dos fármacos , Potássio/urina , Renina/sangue , Sódio/urina , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Membro 1 da Família 12 de Carreador de Soluto/genética
15.
Am J Physiol Renal Physiol ; 308(9): F1026-31, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25715987

RESUMO

Afferent (Af-Art) and efferent arterioles resistance regulate glomerular capillary pressure. The nephron regulates Af-Art resistance via: 1) vasoconstrictor tubuloglomerular feedback (TGF), initiated in the macula densa via Na-K-2Cl cotransporters (NKCC2) and 2) vasodilator connecting tubuloglomerular feedback (CTGF), initiated in connecting tubules via epithelial Na channels (ENaC). Furosemide inhibits NKCC2 and TGF. Benzamil inhibits ENaC and CTGF. In vitro, CTGF dilates preconstricted Af-Arts. In vivo, benzamil decreases stop-flow pressure (PSF), suggesting that CTGF antagonizes TGF; however, even when TGF is blocked, CTGF does not increase PSF, suggesting there is another mechanism antagonizing CTGF. We hypothesize that in addition to NKCC2, activation of Na/H exchanger (NHE) antagonizes CTGF, and when both are blocked CTGF dilates Af-Arts and this effect is blocked by a CTGF inhibitor benzamil. Using micropuncture, we studied the effects of transport inhibitors on TGF responses by measuring PSF while increasing nephron perfusion from 0 to 40 nl/min. Control TGF response (-7.9 ± 0.2 mmHg) was blocked by furosemide (-0.4 ± 0.2 mmHg; P < 0.001). Benzamil restored TGF in the presence of furosemide (furosemide: -0.2 ± 0.1 vs. furosemide+benzamil: -4.3 ± 0.3 mmHg; P < 0.001). With furosemide and NHE inhibitor, dimethylamiloride (DMA), increase in tubular flow increased PSF (furosemide+DMA: 2.7 ± 0.5 mmHg, n = 6), and benzamil blocked this (furosemide+DMA+benzamil: -1.1 ± 0.2 mmHg; P < 0.01, n = 6). We conclude that NHE in the nephron decreases PSF (Af-Art constriction) when NKCC2 and ENaC are inhibited, suggesting that in the absence of NKCC2, NHE causes a TGF response and that CTGF dilates the Af-Art when TGF is blocked with NKCC2 and NHE inhibitors.


Assuntos
Amilorida/análogos & derivados , Bloqueadores do Canal de Sódio Epitelial/farmacologia , Canais Epiteliais de Sódio/efeitos dos fármacos , Furosemida/farmacologia , Glomérulos Renais/efeitos dos fármacos , Túbulos Renais/efeitos dos fármacos , Néfrons/efeitos dos fármacos , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Trocadores de Sódio-Hidrogênio/antagonistas & inibidores , Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Amilorida/farmacologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/fisiologia , Canais Epiteliais de Sódio/metabolismo , Retroalimentação , Glomérulos Renais/irrigação sanguínea , Glomérulos Renais/metabolismo , Túbulos Renais/irrigação sanguínea , Túbulos Renais/metabolismo , Masculino , Néfrons/metabolismo , Ratos Sprague-Dawley , Circulação Renal/efeitos dos fármacos , Trocadores de Sódio-Hidrogênio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Fatores de Tempo , Vasoconstrição/efeitos dos fármacos , Vasodilatação/efeitos dos fármacos
16.
Pflugers Arch ; 467(11): 2325-35, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25603963

RESUMO

GABAA receptor-mediated inhibition--which is due to Cl(-) and HCO3 (-) currents controlled by KCC2 and carbonic anhydrase activity, respectively--contributes to short- and long-lasting interictal events recorded from the CA3 region of hippocampus during application of 4-aminopyridine (4AP, 50 µM). Here, we employed field potential recordings in an in vitro brain slice preparation to establish the effects induced by the KCC2 blockers VU0240551 (10 µM) or bumetanide (50 µM) and by the carbonic anhydrase inhibitor acetazolamide (10 µM) on the two types of interictal events. We found that blocking KCC2 activity decreased the amplitude of the short-lasting events. In addition, this pharmacological procedure increased the interval of occurrence of the long-lasting events and reduced their amplitude. Blocking carbonic anhydrase activity with acetazolamide reduced the interval of occurrence and the duration of the short-lasting events while increasing their amplitude; acetazolamide also reduced the duration and amplitude of the long-lasting events. Finally, blocking either KCC2 or carbonic anhydrase activity increased the interval of occurrence of pharmacologically isolated synchronous GABAergic events and decreased their duration and amplitude. These data substantiate further the role of GABAA receptor-mediated signaling in driving neuronal populations toward hypersynchronous states presumably by increasing extracellular [K(+)].


Assuntos
Anidrases Carbônicas/metabolismo , Convulsões/fisiopatologia , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Acetazolamida/farmacologia , Animais , Região CA3 Hipocampal/efeitos dos fármacos , Inibidores da Anidrase Carbônica/farmacologia , Masculino , Potenciais da Membrana/efeitos dos fármacos , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores de GABA-A/efeitos dos fármacos , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Tiazóis/farmacologia , Tioglicolatos/farmacologia , Ácido gama-Aminobutírico/metabolismo
17.
Am J Physiol Regul Integr Comp Physiol ; 307(6): R653-63, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-24990857

RESUMO

Guanylin (GN) inhibited water absorption and short-circuit current (Isc) in seawater eel intestine. Similar inhibition was observed after bumetanide, and the effect of bumetanide was abolished by GN or vice versa, suggesting that both act on the same target, Na(+)-K(+)-2Cl(-) cotransporter (NKCC), which is a key player for the Na(+)-K(+)-Cl(-) transport system responsible for water absorption in marine teleost intestine. However, effect of GN was always greater than that of bumetanide: 10% greater in middle intestine (MI) and 40% in posterior intestine (PI) for Isc, and 25% greater in MI and 34% in PI for water absorption. After treatment with GN, Isc decreased to zero, but 20-30% water absorption still remained. The remainder may be due to the Cl(-)/HCO3 (-) exchanger and Na(+)-Cl(-) cotransporter (NCC), since inhibitors for these transporters almost nullified the remaining water absorption. Quantitative PCR analysis revealed the presence of major proteins involved in water absorption; the NKCC2ß and AQP1 genes whose expression was markedly upregulated after seawater acclimation. The SLC26A6 (anion exchanger) and NCCß genes were also expressed in small amounts. Consistent with the inhibitors' effect, expression of NKCC2ß was MI > PI, and that of NCCß was MI << PI. The present study showed that GN not only inhibits the bumetanide-sensitive Na(+)-K(+)-Cl(-) transport system governed by NKCC2ß, but also regulates unknown ion transporters different from GN-insensitive SLC26A6 and NCC. A candidate is cystic fibrosis transmembrane conductance regulator Cl(-) channel, as demonstrated in mammals, but its expression is low in eel intestine, and its role may be minor, as indicated by the small effect of its inhibitors.


Assuntos
Enguias/metabolismo , Proteínas de Peixes/antagonistas & inibidores , Hormônios Gastrointestinais/farmacologia , Intestinos/efeitos dos fármacos , Peptídeos Natriuréticos/farmacologia , Água do Mar , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Membro 1 da Família 12 de Carreador de Soluto/antagonistas & inibidores , Água/metabolismo , Adaptação Fisiológica , Animais , Aquaporina 1/genética , Aquaporina 1/metabolismo , Bumetanida/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Enguias/genética , Proteínas de Peixes/genética , Proteínas de Peixes/metabolismo , Mucosa Intestinal/metabolismo , Transporte de Íons , Cinética , Potenciais da Membrana , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Membro 3 da Família 12 de Carreador de Soluto/efeitos dos fármacos , Membro 3 da Família 12 de Carreador de Soluto/genética , Membro 3 da Família 12 de Carreador de Soluto/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...