Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
medRxiv ; 2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37961671

RESUMO

Background: Acute kidney injury (AKI) is common in hospitalized patients with SARS-CoV2 infection despite vaccination and leads to long-term kidney dysfunction. However, peripheral blood molecular signatures in AKI from COVID-19 and their association with long-term kidney dysfunction are yet unexplored. Methods: In patients hospitalized with SARS-CoV2, we performed bulk RNA sequencing using peripheral blood mononuclear cells(PBMCs). We applied linear models accounting for technical and biological variability on RNA-Seq data accounting for false discovery rate (FDR) and compared functional enrichment and pathway results to a historical sepsis-AKI cohort. Finally, we evaluated the association of these signatures with long-term trends in kidney function. Results: Of 283 patients, 106 had AKI. After adjustment for sex, age, mechanical ventilation, and chronic kidney disease (CKD), we identified 2635 significant differential gene expressions at FDR<0.05. Top canonical pathways were EIF2 signaling, oxidative phosphorylation, mTOR signaling, and Th17 signaling, indicating mitochondrial dysfunction and endoplasmic reticulum (ER) stress. Comparison with sepsis associated AKI showed considerable overlap of key pathways (48.14%). Using follow-up estimated glomerular filtration rate (eGFR) measurements from 115 patients, we identified 164/2635 (6.2%) of the significantly differentiated genes associated with overall decrease in long-term kidney function. The strongest associations were 'autophagy', 'renal impairment via fibrosis', and 'cardiac structure and function'. Conclusions: We show that AKI in SARS-CoV2 is a multifactorial process with mitochondrial dysfunction driven by ER stress whereas long-term kidney function decline is associated with cardiac structure and function and immune dysregulation. Functional overlap with sepsis-AKI also highlights common signatures, indicating generalizability in therapeutic approaches. SIGNIFICANCE STATEMENT: Peripheral transcriptomic findings in acute and long-term kidney dysfunction after hospitalization for SARS-CoV2 infection are unclear. We evaluated peripheral blood molecular signatures in AKI from COVID-19 (COVID-AKI) and their association with long-term kidney dysfunction using the largest hospitalized cohort with transcriptomic data. Analysis of 283 hospitalized patients of whom 37% had AKI, highlighted the contribution of mitochondrial dysfunction driven by endoplasmic reticulum stress in the acute stages. Subsequently, long-term kidney function decline exhibits significant associations with markers of cardiac structure and function and immune mediated dysregulation. There were similar biomolecular signatures in other inflammatory states, such as sepsis. This enhances the potential for repurposing and generalizability in therapeutic approaches.

2.
Kidney Med ; 5(2): 100582, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36712313

RESUMO

Rationale & Objective: The association between cannabis use and chronic kidney disease (CKD) is controversial. We aimed to assess association of CKD with cannabis use in a large cohort study and then assess causality using Mendelian randomization with a genome-wide association study (GWAS). Study Design: Retrospective cohort study and genome-wide association study. Setting & Participants: The retrospective study was conducted on the All of Us cohort (N=223,354). Genetic instruments for cannabis use disorder were identified from 3 GWAS: the Psychiatric Genomics Consortium Substance Use Disorders, iPSYCH, and deCODE (N=384,032). Association between genetic instruments and CKD was investigated in the CKDGen GWAS (N > 1.2 million). Exposure: Cannabis consumption. Outcomes: CKD outcomes included: cystatin-C and creatinine-based kidney function, proteinuria, and blood urea nitrogen. Analytical Approach: We conducted association analyses to test for frequency of cannabis use and CKD. To evaluate causality, we performed a 2-sample Mendelian randomization. Results: In the retrospective study, compared to former users, less than monthly (OR, 1.01; 95% CI, 0.87-1.18; P = 0.87) and monthly cannabis users (OR, 1.15; 95% CI, 0.86-1.52; P = 0.33) did not have higher CKD odds. Conversely, weekly (OR, 1.28; 95% CI, 1.01-1.60; P = 0.04) and daily use (OR, 1.25; 95% CI, 1.04-1.50; P = 0.02) was significantly associated with CKD, adjusted for multiple confounders. In Mendelian randomization, genetic liability to cannabis use disorder was not associated with increased odds for CKD (OR, 1.00; 95% CI, 0.99-1.01; P = 0.96). These results were robust across different Mendelian randomization techniques and multiple kidney traits. Limitations: Likely underreporting of cannabis use. In Mendelian randomization, genetic instruments were identified in the GWAS that included individuals primarily of European ancestry. Conclusions: Despite the epidemiological association between cannabis use and CKD, there was no evidence of a causal effect, indicating confounding in observational studies.

4.
J Mol Cell Biol ; 8(6): 464-476, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27190311

RESUMO

Glycogen synthase kinase 3ß (GSK3ß), a serine/threonine protein kinase, is commonly known to be regulated at the level of its activity. However, in some diseases including polycystic kidney disease (PKD), GSK3ß expression is increased and plays a pathophysiological role. The current studies aimed to determine the mechanism for the increased GSK3ß expression in PKD and its significance to disease progression. In mouse models of PKD, increases in renal GSK3ß corresponded with increases in renal cAMP levels and disease progression. In vivo and in vitro studies revealed that GSK3ß is a cAMP-responsive gene, and elevated cAMP levels, as seen in PKD, can increase GSK3ß expression. In normal mice, vasopressin signaling induced by water deprivation increased GSK3ß expression, which decreased following rehydration. Examination of the GSK3ß promoter revealed five potential binding sites for the transcription factor, cAMP response element binding protein (CREB). CREB was found to bind to GSK3ß promoter and essential for cAMP-mediated regulation of GSK3ß. Importantly, this regulation was demonstrated to be part of a feed-forward loop in which cAMP through CREB regulates GSK3ß expression, and GSK3ß in turn positively regulates cAMP generation. GSK3ß or CREB inhibition reduced transepithelial fluid secretion and cyst expansion in vitro Thus, disruption at any point of this destructive cycle may be therapeutically useful to reduce cyst expansion and preserve renal function in PKD.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Doenças Renais Policísticas/metabolismo , Animais , Líquidos Corporais/metabolismo , AMP Cíclico , Cães , Técnicas de Inativação de Genes , Glicogênio Sintase Quinase 3 beta/genética , Humanos , Rim/enzimologia , Rim/patologia , Células Madin Darby de Rim Canino , Camundongos Endogâmicos C57BL , Doenças Renais Policísticas/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais , Canais de Cátion TRPP , Vasopressinas/metabolismo
5.
Curr Opin Nephrol Hypertens ; 24(5): 444-9, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26125646

RESUMO

PURPOSE OF REVIEW: Renal collecting ducts maintain NaCl homeostasis by fine-tuning urinary excretion to balance dietary salt intake. This review focuses on recent studies on transcellular Cl secretion by collecting ducts, its regulation and its role in cyst growth in autosomal dominant polycystic kidney disease (ADPKD). RECENT FINDINGS: Lumens of nonperfused rat medullary collecting ducts collapse in control media but expand with fluid following treatment with cAMP, demonstrating the capacity for both salt absorption and secretion. Recently, inhibition of apical epithelial Na channels (ENaC) unmasked Cl secretion in perfused mouse cortical collecting ducts (CCDs), involving Cl uptake by basolateral NKCC1 and efflux through apical Cl channels. AVP, the key hormone for osmoregulation, promotes cystic fibrosis transmembrane conductance regulator (CFTR)-mediated Cl secretion. In addition, prostaglandin E2 stimulates Cl secretion through both CFTR and Ca-activated Cl channels. SUMMARY: Renal Cl secretion has been commonly overlooked because of the overwhelming capacity for the nephron to reabsorb NaCl from the glomerular filtrate. In ADPKD, Cl secretion plays a central role in the accumulation of cyst fluid and the remarkable size of the cystic kidneys. Investigation of renal Cl secretion may provide a better understanding of NaCl homeostasis and identify new approaches to reduce cyst growth in PKD.


Assuntos
Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Sódio/metabolismo , Animais , Humanos , Néfrons/metabolismo
6.
Kidney Int ; 87(6): 1164-75, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25629553

RESUMO

Polycystic kidney diseases (PKDs) are inherited disorders characterized by the formation of fluid filled renal cysts. Elevated cAMP levels in PKDs stimulate progressive cyst enlargement involving cell proliferation and transepithelial fluid secretion often leading to end-stage renal disease. The glycogen synthase kinase-3 (GSK3) family of protein kinases consists of GSK3α and GSK3ß isoforms and has a crucial role in multiple cellular signaling pathways. We previously found that GSK3ß, a regulator of cell proliferation, is also crucial for cAMP generation and vasopressin-mediated urine concentration by the kidneys. However, the role of GSK3ß in the pathogenesis of PKDs is not known. Here we found that GSK3ß expression and activity were markedly upregulated and associated with cyst-lining epithelia in the kidneys of mice and humans with PKD. Renal collecting duct-specific gene knockout of GSK3ß or pharmacological inhibition of GSK3 effectively slowed down the progression of PKD in mouse models of autosomal recessive or autosomal dominant PKD. GSK3 inactivation inhibited cAMP generation and cell proliferation resulting in reduced cyst expansion, improved renal function, and extended life span. GSK3ß inhibition also reduced pERK, c-Myc, and cyclin-D1, known mitogens in proliferation of cystic epithelial cells. Thus, GSK3ß has a novel functional role in PKD pathophysiology, and its inhibition may be therapeutically useful to slow down cyst expansion and progression of PKD.


Assuntos
AMP Cíclico/metabolismo , Cistos/metabolismo , Cistos/patologia , Quinase 3 da Glicogênio Sintase/metabolismo , Doenças Renais Policísticas/enzimologia , Animais , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Humanos , Rim/enzimologia , Túbulos Renais Coletores/enzimologia , Camundongos , Camundongos Knockout , Tamanho do Órgão/efeitos dos fármacos , Doenças Renais Policísticas/patologia , Doenças Renais Policísticas/fisiopatologia , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Mensageiro/metabolismo , Tiadiazóis/farmacologia
7.
J Biol Chem ; 289(10): 6475-6484, 2014 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-24436330

RESUMO

The first extracellular loop (ECL1) of claudins forms paracellular pores in the tight junction that determine ion permselectivity. We aimed to map the pore-lining residues of claudin-2 by comprehensive cysteine-scanning mutagenesis of ECL1. We screened 45 cysteine mutations within the ECL1 by expression in polyclonal Madin-Darby canine kidney II Tet-Off cells and found nine mutants that displayed a significant decrease of conductance after treatment with the thiol-reactive reagent 2-(trimethylammonium)ethyl methanethiosulfonate, indicating the location of candidate pore-lining residues. Next, we stably expressed these candidates in monoclonal Madin-Darby canine kidney I Tet-Off cells and exposed them to thiol-reactive reagents. The maximum degree of inhibition of conductance, size selectivity of degree of inhibition, and size dependence of the kinetics of reaction were used to deduce the location of residues within the pore. Our data support the following sequence of pore-lining residues located from the narrowest to the widest part of the pore: Ser(68), Ser(47), Thr(62)/Ile(66), Thr(56), Thr(32)/Gly(45), and Met(52). The paracellular pore appears to primarily be lined by polar side chains, as expected for a predominantly aqueous environment. Furthermore, our results strongly suggest the existence of a continuous sequence of residues in the ECL1 centered around Asp(65)-Ser(68) that form a major part of the lining of the pore.


Assuntos
Claudinas/metabolismo , Cisteína/metabolismo , Epitélio/metabolismo , Sequência de Aminoácidos , Animais , Claudinas/química , Claudinas/genética , Cisteína/química , Cisteína/genética , Cães , Epitélio/ultraestrutura , Testes Genéticos , Células Madin Darby de Rim Canino , Mesilatos/química , Camundongos , Dados de Sequência Molecular , Mutagênese , Porosidade , Estrutura Secundária de Proteína
8.
Am J Physiol Cell Physiol ; 306(3): C263-78, 2014 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-24284792

RESUMO

Under conditions of high dietary salt intake, prostaglandin E2 (PGE2) production is increased in the collecting duct and promotes urinary sodium chloride (NaCl) excretion; however, the molecular mechanisms by which PGE2 increases NaCl excretion in this context have not been clearly defined. We used the mouse inner medullary collecting duct (mIMCD)-K2 cell line to characterize mechanisms underlying PGE2-regulated NaCl transport. When epithelial Na(+) channels were inhibited, PGE2 exclusively stimulated basolateral EP4 receptors to increase short-circuit current (Isc(PGE2)). We found that Isc(PGE2) was sensitive to inhibition by H-89 and CFTR-172, indicating that EP4 receptors signal through protein kinase A to induce Cl(-) secretion via cystic fibrosis transmembrane conductance regulator (CFTR). Unexpectedly, we also found that Isc(PGE2) was sensitive to inhibition by BAPTA-AM (Ca(2+) chelator), 2-aminoethoxydiphenyl borate (2-APB) (inositol triphosphate receptor blocker), and flufenamic acid (FFA) [Ca(2+)-activated Cl(-) channel (CACC) inhibitor], suggesting that EP4 receptors also signal through Ca(2+) to induce Cl(-) secretion via CACC. Additionally, we observed that PGE2 stimulated an increase in Isc through crosstalk between cAMP and Ca(2+) signaling; BAPTA-AM or 2-APB inhibited a component of Isc(PGE2) that was sensitive to CFTR-172 inhibition; H-89 inhibited a component of Isc(PGE2) that was sensitive to FFA inhibition. Together, our findings indicate that PGE2 activates basolateral EP4 receptors and signals through both cAMP and Ca(2+) to stimulate Cl(-) secretion in IMCD-K2 cells. We propose that these signaling pathways, and the crosstalk between them, may provide a concerted mechanism for enhancing urinary NaCl excretion under conditions of high dietary NaCl intake.


Assuntos
Canais de Cloreto/metabolismo , AMP Cíclico/metabolismo , Dinoprostona/metabolismo , Canais de Sódio/metabolismo , Cloreto de Sódio/metabolismo , Animais , Benzoatos/farmacologia , Compostos de Boro , Cálcio/metabolismo , Sinalização do Cálcio , Linhagem Celular , Canais de Cloreto/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Ácido Egtázico/análogos & derivados , Ácido Egtázico/farmacologia , Ácido Flufenâmico/farmacologia , Receptores de Inositol 1,4,5-Trifosfato/antagonistas & inibidores , Transporte de Íons , Isoquinolinas/farmacologia , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Camundongos , Técnicas de Patch-Clamp , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Bloqueadores dos Canais de Sódio , Sulfonamidas/farmacologia , Tiazolidinas/farmacologia
9.
Annu Rev Physiol ; 75: 479-501, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23140368

RESUMO

Claudins are tight junction membrane proteins that regulate paracellular permeability of renal epithelia to small ions, solutes, and water. Claudins interact within the cell membrane and between neighboring cells to form tight junction strands and constitute both the paracellular barrier and the pore. The first extracellular domain of claudins is thought to be the pore-lining domain and contains the determinants of charge selectivity. Multiple claudins are expressed in different nephron segments; such differential expression likely determines the permeability properties of each segment. Recent evidence has identified claudin-2 as constituting the cation-reabsorptive pathway in the proximal tubule; claudin-14, -16, and -19 as forming a complex that regulates calcium transport in the thick ascending limb of the loop of Henle; and claudin-4, -7, and -8 as determinants of collecting duct chloride permeability. Mutations in claudin-16 and -19 cause familial hypercalciuric hypomagnesemia with nephrocalcinosis. The roles of other claudins in kidney diseases remain to be fully elucidated.


Assuntos
Claudinas/fisiologia , Rim/fisiologia , Junções Íntimas/fisiologia , Animais , Transporte Biológico/fisiologia , Cálcio/metabolismo , Permeabilidade da Membrana Celular/fisiologia , Humanos , Rim/citologia , Nefropatias/patologia , Nefropatias/fisiopatologia
10.
Am J Physiol Renal Physiol ; 303(10): F1425-34, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-22933297

RESUMO

Prostaglandin E(2) (PGE(2)) contributes to cystogenesis in genetically nonorthologous models of autosomal dominant polycystic kidney disease (ADPKD). However, it remains unknown whether PGE(2) induces the classic features of cystic epithelia in genetically orthologous models of ADPKD. We hypothesized that, in ADPKD epithelia, PGE(2) induces proliferation and chloride (Cl(-)) secretion, two archetypal phenotypic features of ADPKD. To test this hypothesis, proliferation and Cl(-) secretion were measured in renal epithelial cells deficient in polycystin-1 (PC-1). PC-1-deficient cells increased in cell number (proliferated) faster than PC-1-replete cells, and this proliferative advantage was abrogated by cyclooxygenase inhibition, indicating a role for PGE(2) in cell proliferation. Exogenous administration of PGE(2) increased proliferation of PC-1-deficient cells by 38.8 ± 5.2% (P < 0.05) but inhibited the growth of PC-1-replete control cells by 49.4 ± 1.9% (P < 0.05). Next, we tested whether PGE(2)-specific E prostanoid (EP) receptor agonists induce intracellular cAMP and downstream ß-catenin activation. PGE(2) and EP4 receptor agonism (TCS 2510) increased intracellular cAMP concentration and the abundance of active ß-catenin in PC-1-deficient cells, suggesting a mechanism for PGE(2)-mediated proliferation. Consistent with this hypothesis, antagonizing EP4 receptors reverted the growth advantage of PC-1-deficient cells, implicating a central role for the EP4 receptor in proliferation. To test whether PGE(2)-dependent Cl(-) secretion is also enhanced in PC-1-deficient cells, we used an Ussing chamber to measure short-circuit current (I(sc)). Addition of PGE(2) induced a fivefold higher increase in I(sc) in PC-1-deficient cells compared with PC-1-replete cells. This PGE(2)-induced increase in I(sc) in PC-1-deficient cells was blocked by CFTR-172 and flufenamic acid, indicating that PGE(2) activates CFTR and calcium-activated Cl(-) channels. In conclusion, PGE(2) activates aberrant signaling pathways in PC-1-deficient epithelia that contribute to the proliferative and secretory phenotype characteristic of ADPKD and suggests a therapeutic role for PGE(2) inhibition and EP4 receptor antagonism.


Assuntos
Proliferação de Células/efeitos dos fármacos , Cloretos/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/metabolismo , Rim/metabolismo , Rim Policístico Autossômico Dominante/metabolismo , Canais de Cátion TRPP/metabolismo , Animais , Linhagem Celular , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Rim/efeitos dos fármacos , Camundongos , Rim Policístico Autossômico Dominante/genética , Canais de Cátion TRPP/genética
11.
Am J Physiol Renal Physiol ; 303(4): F483-91, 2012 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-22647633

RESUMO

Extracellular ATP in the cortical collecting duct can inhibit epithelial sodium channels (ENaC) but also stimulate calcium-activated chloride channels (CACC). The relationship between ATP-mediated regulation of ENaC and CACC activity in cortical collecting duct cells has not been clearly defined. We used the mpkCCD(c14) cortical collecting duct cell line to determine effects of ATP on sodium (Na(+)) and chloride (Cl(-)) transport with an Ussing chamber system. ATP, at a concentration of 10(-6) M or less, did not inhibit ENaC-mediated short-circuit current (I(sc)) but instead stimulated a transient increase in I(sc). The macroscopic current-voltage relationship for ATP-inducible current demonstrated that the direction of this ATP response changes from positive to negative when transepithelial voltage (V(te)) is clamped to less than -10 mV. We hypothesized that this negative V(te) might be found under conditions of aldosterone stimulation. We next stimulated mpkCCD(c14) cells with aldosterone (10(-6) M) and then clamped the V(te) to -50 mV, the V(te) of aldosterone-stimulated cells under open-circuit conditions. ATP (10(-6) M) induced a transient increase in negative clamp current, which could be inhibited by flufenamic acid (CACC inhibitor) and BAPTA-AM (calcium chelator), suggesting that ATP stimulates Cl(-) absorption through CACC. Together, our findings suggest that the status of ENaC activity, by controlling V(te), may dictate the direction of ATP-stimulated Cl(-) transport. This interplay between aldosterone and purinergic signaling pathways may be relevant for regulating NaCl transport in cortical collecting duct cells under different states of extracellular fluid volume.


Assuntos
Trifosfato de Adenosina/metabolismo , Cloretos/metabolismo , Córtex Renal/citologia , Túbulos Renais Coletores/citologia , Absorciometria de Fóton , Trifosfato de Adenosina/genética , Animais , Anoctamina-1 , Bestrofinas , Transporte Biológico/fisiologia , Linhagem Celular , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Regulação da Expressão Gênica , Camundongos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Receptores Purinérgicos P2Y/metabolismo
12.
J Biol Chem ; 286(37): 32074-85, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21784856

RESUMO

Serum- and glucocorticoid-regulated kinase 1 (sgk1) participates in diverse biological processes, including cell growth, apoptosis, and sodium homeostasis. In the cortical collecting duct of the kidney, sgk1 regulates sodium transport by stimulating the epithelial sodium channel (ENaC). Control of subcellular localization of sgk1 may be an important mechanism for modulating specificity of sgk1 function; however, which subcellular locations are required for sgk1-regulated ENaC activity in collecting duct cells has yet to be established. Using cell surface biotinylation studies, we detected endogenous sgk1 at the apical cell membrane of aldosterone-stimulated mpkCCD(c14) collecting duct cells. The association of sgk1 with the cell membrane was enhanced when ENaC was co-transfected with sgk1 in kidney cells, suggesting that ENaC brings sgk1 to the cell surface. Furthermore, association of endogenous sgk1 with the apical cell membrane of mpkCCD(c14) cells could be modulated by treatments that increase or decrease ENaC expression at the apical membrane; forskolin increased the association of sgk1 with the apical surface, whereas methyl-ß-cyclodextrin decreased the association of sgk1 with the apical surface. Single channel recordings of excised inside-out patches from the apical membrane of aldosterone-stimulated A6 collecting duct cells revealed that the open probability of ENaC was sensitive to the sgk1 inhibitor GSK650394, indicating that endogenous sgk1 is functionally active at the apical cell membrane. We propose that the association of sgk1 with the apical cell membrane, where it interacts with ENaC, is a novel means by which sgk1 specifically enhances ENaC activity in aldosterone-stimulated collecting duct cells.


Assuntos
Membrana Celular/metabolismo , Canais Epiteliais de Sódio/metabolismo , Proteínas Imediatamente Precoces/metabolismo , Túbulos Renais Coletores/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Aldosterona/farmacologia , Animais , Benzoatos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Membrana Celular/genética , Canais Epiteliais de Sódio/genética , Células HEK293 , Humanos , Proteínas Imediatamente Precoces/antagonistas & inibidores , Proteínas Imediatamente Precoces/genética , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/fisiologia , Camundongos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Sódio/metabolismo , beta-Ciclodextrinas/farmacologia
13.
Am J Physiol Renal Physiol ; 301(3): F544-53, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21653634

RESUMO

Dysregulation of urinary sodium chloride (NaCl) excretion can result in extracellular fluid (ECF) volume expansion and hypertension. Recent studies demonstrated that urinary nucleotide excretion increases in mice ingesting a high-salt diet and that these increases in extracellular nucleotides can signal through P2Y(2) receptors in the kidney collecting duct to inhibit epithelial Na(+) channels (ENaC). However, under conditions of ECF volume expansion brought about by high-dietary salt intake, ENaC activity should already be suppressed. We hypothesized that alternative pathways exist by which extracellular nucleotides control renal NaCl excretion. We used an inner medullary collecting duct (mIMCD-K2) cell line in an Ussing chamber system as a model to study additional ion transport pathways that are regulated by extracellular nucleotides. When ENaC was inhibited, the addition of adenosine triphosphate (ATP) to the basal side of cell sheets activated both P2Y(1) and P2Y(2) receptors, inducing a transient increase in short-circuit current (I(sc)); addition of ATP to the apical side activated only P2Y(2) receptors, inducing first a transient and then a sustained increase in I(sc). The ATP-induced increases in I(sc) were blocked by pretreatment with a phospholipase C (PLC) inhibitor, a calcium (Ca(2+)) chelator, or Ca(2+)-activated Cl(-) channel (CACC) inhibitors, suggesting that ATP signals through both PLC and intracellular Ca(2+) to activate CACC. We propose that P2Y(1) and P2Y(2) receptors operate in tandem in IMCD cells to provide an adaptive mechanism for enhancing urinary NaCl excretion in the setting of high-dietary NaCl intake.


Assuntos
Canais de Cloreto/metabolismo , Cloretos/metabolismo , Túbulos Renais Coletores/citologia , Túbulos Renais Coletores/metabolismo , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2Y2/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Cálcio/metabolismo , Linhagem Celular , Transporte de Íons/fisiologia , Camundongos , Modelos Animais , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Cloreto de Sódio/metabolismo , Cloreto de Sódio na Dieta/farmacologia , Fosfolipases Tipo C/metabolismo
14.
Peptides ; 32(5): 1068-72, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21335042

RESUMO

Gamma-Melanocyte Stimulating Hormone (Gamma-MSH) regulates sodium (Na(+)) balance and blood pressure through activation of the melanocortin receptor 3 (MC3-R). The mechanism of the natriuretic effect is proposed to involve binding of MC3-R either in the kidney to directly inhibit tubular Na(+) transport or in the brain to inhibit central neural pathways that control renal tubular Na(+) absorption. This study aimed to clarify the mechanism involved in the natriuretic effect of Gamma-MSH on MC3-R in kidney cells. In Ussing chamber studies, we observed no effects of Gamma-MSH on NaCl transport in the mouse inner medullary collecting duct cell line (mIMCD-K2). We also found that neither MC3-R protein nor mRNA was expressed in mouse kidney, suggesting that renal Gamma-MSH action may not be mediated through direct effects on tubular Na(+) transport but rather through effects on central neural pathways that innervate the kidney.


Assuntos
gama-MSH/farmacologia , Animais , Transporte Biológico/efeitos dos fármacos , Western Blotting , Linhagem Celular , AMP Cíclico/metabolismo , Imuno-Histoquímica , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor Tipo 3 de Melanocortina/genética , Receptor Tipo 3 de Melanocortina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Sódio/metabolismo
15.
Hypertension ; 55(5): 1123-8, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20308611

RESUMO

In the kidney, defects in the regulation of urine salt excretion can result in extracellular fluid volume expansion, leading to salt-sensitive hypertension. Previous studies have demonstrated that, when rats are maintained on a high sodium chloride (NaCl) diet, adenosine production increases in the renal medulla with parallel changes in adenosine receptor expression. These studies suggest that adenosine signaling in the kidney can respond to high NaCl loading; however, the functional consequences of these changes in adenosine signaling are not clear. We used the immortalized cell line mIMCD-K2, a murine model system for the renal inner medullary collecting duct, to study the direct effects of adenosine on NaCl transport across the inner medullary collecting duct epithelium with an Ussing chamber system. When epithelial Na(+) channels were inhibited, the addition of adenosine to the apical side of mIMCD-K2 cell sheets stimulated short-circuit current in a dose-dependent manner. This increase in short-circuit current was inhibited by a cystic fibrosis transmembrane conductance regulator Cl(-) channel inhibitor. Pharmacological studies with a panel of adenosine receptor agonists and antagonists demonstrated that adenosine activates apical A2b adenosine receptors to enhance the short-circuit current. Furthermore, adenosine application to mIMCD-K2 cell sheets increased intracellular cAMP, whereas inhibition of protein kinase A completely blocked the adenosine response. Together, our findings indicate that adenosine stimulates Cl(-) secretion through the cystic fibrosis transmembrane conductance regulator in mIMCD-K2 cells by activating apical A2b receptors and signaling through cAMP/protein kinase A. We propose that this adenosine receptor pathway may provide one mechanism for enhancing urine NaCl excretion in the setting of high dietary NaCl intake.


Assuntos
Adenosina/farmacologia , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Medula Renal/metabolismo , Túbulos Renais Coletores/metabolismo , Receptor A2B de Adenosina/fisiologia , Animais , Linhagem Celular , Regulador de Condutância Transmembrana em Fibrose Cística/efeitos dos fármacos , Medula Renal/efeitos dos fármacos , Túbulos Renais Coletores/efeitos dos fármacos , Pirazóis/farmacologia , Pirimidinas/farmacologia , Ratos , Receptor A2B de Adenosina/efeitos dos fármacos , Triazinas/farmacologia , Triazóis/farmacologia , Xantinas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA