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











Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Renal Physiol ; 326(1): F152-F164, 2024 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-37969102

RESUMO

As miR-137 is a regulator of aquaporin (AQP)2 expression and tumor necrosis factor (TNF) inhibits the expression of several extrarenal AQPs, we tested the hypothesis that TNF inhibits AQP2 in the kidney via a miR-137-dependent mechanism. AQP2 mRNA and protein expression decreased ∼70% and 53%, respectively, in primary renal inner medullary collecting duct (IMCD) cells transfected with a miRNA mimic of mmu-miR-137, suggesting that miR-137 directly targets AQP2 mRNA in these cells. Exposure of IMCD cells for 2 h to 400 mosmol/kgH2O medium increased mmu-miR-137 mRNA expression about twofold, conditions that also increased TNF production approximately fourfold. To determine if the increase in mmu-miR-137 mRNA expression was related to the concomitant increase in TNF, IMCD cells were transfected with a lentivirus construct to silence TNF. This construct decreased mmu-miR-137 mRNA expression by ∼63%, suggesting that TNF upregulates the expression of miR-137. Levels of miR-137 also increased approximately twofold in IMCD tubules isolated from male mice given 1% NaCl in the drinking water for 3 days. Intrarenal lentivirus silencing of TNF increased AQP2 mRNA levels and protein expression concomitant with a decrease in miR-137 levels in tubules isolated from mice given NaCl. The changes in AQP2 expression levels affected the diluting ability of the kidney, which was assessed by measuring urine osmolality and urine volume, as the decrease in these parameters after renal silencing of TNF was prevented on intrarenal administration of miR-137. The study reveals a novel TNF function via a miR-137-dependent mechanism that regulates AQP2 expression and function.NEW & NOTEWORTHY An emerging intratubular tumor necrosis factor system, functioning during normotensive noninflammatory conditions, acts as a breaking mechanism that attenuates both the increases in Na+-K+-2Cl- cotransporter and aquaporin-2 induced by arginine vasopressin, thereby contributing to the regulation of electrolyte balance and blood pressure. A greater appreciation for the role of cytokines as mediators of immunophysiological responses may help reveal the relationship between the immune system and other physiological systems.


Assuntos
Aquaporinas , Túbulos Renais Coletores , MicroRNAs , Camundongos , Masculino , Animais , Aquaporina 2/genética , Aquaporina 2/metabolismo , Cloreto de Sódio/metabolismo , Túbulos Renais Coletores/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Aquaporinas/metabolismo
2.
Hypertension ; 80(2): 426-439, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36448465

RESUMO

BACKGROUND: Previous studies showed that miR-195a-5p was among the most abundant microRNAs (miRNAs) expressed in the kidney. METHODS: Lentivirus silencing of tumor necrosis factor-α (TNF) was performed in vivo and in vitro. Luciferase reporter assays confirmed that bumetanide-sensitive Na+-K+-2Cl- cotransporter isoform A (NKCC2A) mRNA is targeted and repressed by miR-195a-5p. Radiotelemetry was used to measure mean arterial pressure. RESULTS: TNF upregulates mmu-miR-195a-5p, and -203 and downregulates mmu-miR-30c and -100 in the medullary thick ascending limb of male mice. miR-195a-5p was >3-fold higher in the renal outer medulla of mice given an intrarenal injection of murine recombinant TNF, whereas silencing TNF inhibited miR-195a-5p expression by ≈51%. Transient transfection of a miR-195a-5p mimic into medullary thick ascending limb cells suppressed NKCC2A mRNA by ≈83%, whereas transfection with Anti-miR-195a-5p increased NKCC2A mRNA. Silencing TNF in medullary thick ascending limb cells prevented increases in miR-195 induced by 400 mosmol/kg H2O medium, an effect reversed by transfection with a miR-195a-5p mimic. Expression of phosphorylated NKCC2 increased 1.5-fold in medullary thick ascending limb cells transfected with Anti-miR-195a-5p and a miR-195a-5p mimic prevented the increase, which was induced by silencing TNF in cells exposed to 400 mosmol/kg H2O medium after osmolality was increased by adding NaCl. Intrarenal injection of TNF suppressed NKCC2A mRNA, whereas injection of miR-195a-5p prevented the increase of NKCC2A mRNA abundance and phosphorylated NKCC2 expression when TNF was silenced. Intrarenal injection with miR-195a-5p markedly attenuated MAP after renal silencing of TNF in mice given 1% NaCl. CONCLUSIONS: The study identifies miR-195a-5p as a salt-sensitive and TNF-inducible miRNA that attenuates NaCl-mediated increases in blood pressure by inhibiting NKCC2A.


Assuntos
MicroRNAs , Cloreto de Sódio , Animais , Masculino , Camundongos , Antagomirs , Pressão Sanguínea/fisiologia , MicroRNAs/genética , MicroRNAs/metabolismo , RNA Mensageiro/genética , Cloreto de Sódio/farmacologia , Cloreto de Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
3.
J Hum Hypertens ; 37(8): 701-708, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-36008598

RESUMO

Previous work in mouse models shows that urinary TNF-α levels become elevated when dietary salt (NaCl) intake increases. To examine if this relationship exists in humans, we conducted a secondary analysis of the Dietary Approaches to Stop Hypertension (DASH)-Sodium trial to determine levels of urinary TNF-α in 367 subjects categorized by race, sex, and blood pressure. The DASH-Sodium trial is a multicenter feeding trial in which subjects were randomly assigned to either the DASH or control diet, and high, medium, and low sodium in random order. Multivariable linear regression was used to model baseline TNF-α and a mixed model was used to model TNF-α as a function of dietary intervention. At baseline, with all subjects on a "typical American diet", urinary TNF-α levels were lowest in Black, p = 0.002 and male subjects, p < 0.001. After randomization to either the DASH or control diet, with increasing levels of sodium, urinary TNF-α levels increased only in subjects on the control diet, p < 0.05. As in the baseline analysis, TNF-α levels were highest in White females, then White males, Black females and lowest in Black males. The results indicate that urinary TNF-α levels in DASH-Sodium subjects are regulated by NaCl intake, modulated by the DASH diet, and influenced by both race and sex. The inherent differences between subgroups support studies in mice showing that increases in renal TNF-α minimize the extent salt-dependent activation of NKCC2.


Assuntos
Abordagens Dietéticas para Conter a Hipertensão , Hipertensão , Sódio na Dieta , Feminino , Humanos , Masculino , Animais , Camundongos , Sódio/urina , Fator de Necrose Tumoral alfa , Cloreto de Sódio , Fatores Raciais , Dieta Hipossódica , Pressão Sanguínea , Cloreto de Sódio na Dieta
4.
Hypertension ; 79(12): 2656-2670, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36129177

RESUMO

TNF-α (tumor necrosis factor-alpha) is the best known as a proinflammatory cytokine; yet, this cytokine also has important immunomodulatory and regulatory functions. As the effects of TNF-α on immune system function were being revealed, the spectrum of its activities appeared in conflict with each other before investigators defined the settings and mechanisms by which TNF-α contributed to both host defense and chronic inflammation. These effects reflect self-protective mechanisms that may become harmful when dysregulated. The paradigm of physiological and pathophysiological effects of TNF-α has since been uncovered in the lung, colon, and kidney where its role has been identified in pulmonary edema, electrolyte reabsorption, and blood pressure regulation, respectively. Recent studies on the prohypertensive and inflammatory effects of TNF-α in the cardiovascular system juxtaposed to those related to NaCl and blood pressure homeostasis, the response of the kidney to lipopolysaccharide, and protection against bacterial infections are helping define the mechanisms by which TNF-α modulates distinct functions within the kidney. This review discusses how production of TNF-α by renal epithelial cells may contribute to regulatory mechanisms that not only govern electrolyte excretion and blood pressure homeostasis but also maintain the appropriate local hypersalinity environment needed for optimizing the innate immune response to bacterial infections in the kidney. It is possible that the wide range of effects mediated by TNF-α may be related to severity of disease, amount of inflammation and TNF-α levels, and the specific cell types that produce this cytokine, areas that remain to be investigated further.


Assuntos
Angiotensina II , Fator de Necrose Tumoral alfa , Humanos , Pressão Sanguínea/fisiologia , Angiotensina II/farmacologia , Fator de Necrose Tumoral alfa/metabolismo , Lipopolissacarídeos/farmacologia , Cloreto de Sódio na Dieta/farmacologia , Rim/metabolismo , Citocinas/metabolismo , Inflamação/metabolismo
5.
Am J Physiol Renal Physiol ; 320(6): F1159-F1164, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33969695

RESUMO

Although administration of hypertonic saline (HSS) in combination with diuretics has yielded improved weight loss, preservation of renal function, and reduction in hospitalization time in the clinical setting of patients with acute decompensated heart failure (ADHF), the mechanisms that underlie these beneficial effects remain unclear and additional studies are needed before this approach can be adopted on a more consistent basis. As high salt conditions stimulate the production of several renal autacoids that exhibit natriuretic effects, renal physiologists can contribute to the understanding of mechanisms by which HSS leads to increased diuresis both as an individual therapy as well as in combination with loop diuretics. For instance, since HSS increases TNF-α production by proximal tubule and thick ascending limb of Henle's loop epithelial cells, this article is aimed at highlighting how the effects of TNF-α produced by these cell types may contribute to the beneficial effects of HSS in patients with ADHF. Although TNF-α produced by infiltrating macrophages and T cells exacerbates and attenuates renal damage, respectively, production of this cytokine within the tubular compartment of the kidney functions as an intrinsic regulator of blood pressure and Na+ homeostasis via mechanisms along the nephron related to inhibition of Na+-K+-2Cl- cotransporter isoform 2 activity and angiotensinogen expression. Thus, in the clinical setting of ADHF and hyponatremia, induction of TNF-α production along the nephron by administration of HSS may attenuate Na+-K+-2Cl- cotransporter isoform 2 activity and angiotensinogen expression as part of a mechanism that prevents excessive Na+ reabsorption in the thick ascending limb of Henle's loop, thereby mitigating volume overload.


Assuntos
Insuficiência Cardíaca/tratamento farmacológico , Solução Salina Hipertônica/farmacologia , Fator de Necrose Tumoral alfa/agonistas , Diuréticos/uso terapêutico , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/genética , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
6.
Hypertension ; 76(6): 1744-1752, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33131307

RESUMO

We showed that intrarenal suppression of TNF (tumor necrosis factor) production under low salt (LS) conditions increases renal cortical AGT (angiotensinogen) mRNA and protein expression. Intrarenal injection of murine recombinant TNF attenuated increases of AGT in mice ingesting LS. Moreover, AGT mRNA and protein expression increased ≈6-fold and 2-fold, respectively, in mice ingesting LS that also received an intrarenal injection of a lentivirus construct that specifically silenced TNF in the kidney (U6-TNF-ex4). Silencing of TNF under normal salt and high salt (HS) conditions also resulted in increased AGT expression. Since renal TNF production decreases in response to LS and increases in response to HS, the data suggest that alterations in TNF production under these conditions modulate the degree of AGT expression. We also tested the hypothesis that TNF inhibits intrarenal AGT expression by a mechanism involving miR-133a. Expression of miR-133a decreased in mice given LS and increased in response to HS for 7 days. Intrarenal silencing of TNF reversed the effects of HS on miR-133a-dependent AGT expression. In contrast, intrarenal TNF administration increased miR-133a expression in the kidney. Collectively, the data suggest that miR-133a is a salt-sensitive microRNA that inhibits AGT in the kidney and is increased by TNF. The HS-induced increase in blood pressure observed following silencing of TNF was markedly reduced upon intrarenal administration of miR-133a suggesting that intrinsic effects of TNF in the kidney to limit the blood pressure response to HS include an increase in miR-133a, which suppresses AGT expression.


Assuntos
Angiotensinogênio/genética , Regulação da Expressão Gênica/genética , Túbulos Renais Proximais/metabolismo , MicroRNAs/genética , Fator de Necrose Tumoral alfa/genética , Angiotensinogênio/metabolismo , Animais , Pressão Sanguínea/efeitos dos fármacos , Pressão Sanguínea/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Túbulos Renais Proximais/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Interferência de RNA , Cloreto de Sódio na Dieta/administração & dosagem , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/metabolismo
7.
Am J Physiol Renal Physiol ; 318(1): F273-F282, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31813248

RESUMO

We have previously shown that TNF-α produced by renal epithelial cells inhibits Na+-K+-2Cl- cotransporter (NKCC2) activity as part of a mechanism that attenuates increases in blood pressure in response to high NaCl intake. As the role of TNF-α in the kidney is still being defined, the effects of low salt intake on TNF-α and NKCC2B expression were determined. Mice given a low-salt (0.02% NaCl) diet (LSD) for 7 days exhibited a 62 ± 7.4% decrease in TNF-α mRNA accumulation in the renal cortex. Mice that ingested the LSD also exhibited an ~63% increase in phosphorylated NKCC2 expression in the cortical thick ascending limb of Henle's loop and a concomitant threefold increase in NKCC2B mRNA abundance without a concurrent change in NKCC2A mRNA accumulation. NKCC2B mRNA levels increased fivefold in mice that ingested the LSD and also received an intrarenal injection of a lentivirus construct that specifically silenced TNF-α in the kidney (U6-TNF-ex4) compared with mice injected with control lentivirus. Administration of a single intrarenal injection of murine recombinant TNF-α (5 ng/g body wt) attenuated the increases of NKCC2B mRNA by ~50% and inhibited the increase in phosphorylated NKCC2 by ~54% in the renal cortex of mice given the LSD for 7 days. Renal silencing of TNF-α decreased urine volume and NaCl excretion in mice given the LSD, effects that were reversed when NKCC2B was silenced in the kidney. Collectively, these findings demonstrate that downregulation of renal TNF-α production in response to low-salt conditions contributes to the regulation of NaCl reabsorption via an NKCC2B-dependent mechanism.


Assuntos
Dieta Hipossódica , Córtex Renal/metabolismo , Cloreto de Sódio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Pressão Sanguínea/fisiologia , Técnicas de Silenciamento de Genes , Alça do Néfron/metabolismo , Camundongos , Fosforilação , Membro 1 da Família 12 de Carreador de Soluto/genética , Fator de Necrose Tumoral alfa/genética
8.
Hypertension ; 71(6): 1117-1125, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29735631

RESUMO

We tested the hypothesis that TNF (tumor necrosis factor)-α produced within the kidney and acting on the renal tubular system is part of a regulatory mechanism that attenuates increases in blood pressure in response to high salt intake. Intrarenal administration of a lentivirus construct, which specifically silenced TNF in the kidney, did not affect baseline blood pressure. However, blood pressure increased significantly 1 day after mice with intrarenal silencing of TNF ingested 1% NaCl in the drinking water. The increase in blood pressure, which was continuously observed for 11 days, promptly returned to baseline levels when mice were switched from 1% NaCl to tap water. Silencing of renal TNF also increased NKCC2 (Na+-K+-2Cl- cotransporter) phosphorylation and induced a selective increase in NKCC2A (NKCC2 isoform A) mRNA accumulation in both the cortical and medullary thick ascending limb of Henle loop that was neither associated with a compensatory decrease of NKCC2F in the medulla nor NKCC2B in the cortex. The NaCl-mediated increases in blood pressure were completely absent when NKCC2A, using a lentivirus construct that did not alter expression of NKCC2F or NKCC2B, and TNF were concomitantly silenced in the kidney. Moreover, the decrease in urine volume and NaCl excretion induced by renal TNF silencing was abolished when NKCC2A was concurrently silenced, suggesting that this isoform contributes to the transition from a salt-resistant to salt-sensitive phenotype. Collectively, the data are the first to demonstrate a role for TNF produced by the kidney in the modulation of sodium homeostasis and blood pressure regulation.


Assuntos
Pressão Sanguínea/fisiologia , Regulação da Expressão Gênica , Hipertensão Renal/genética , Rim/metabolismo , Membro 1 da Família 12 de Carreador de Soluto/genética , Fator de Necrose Tumoral alfa/metabolismo , Animais , Western Blotting , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hipertensão Renal/metabolismo , Hipertensão Renal/fisiopatologia , Rim/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cloreto de Sódio/toxicidade , Membro 1 da Família 12 de Carreador de Soluto/biossíntese , Transcrição Gênica
9.
J Matern Fetal Neonatal Med ; 28(8): 869-75, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25034210

RESUMO

We tested the hypothesis that maternal peripheral blood leukocytes contribute to elevated levels of soluble TNF receptors (sTNFR) in preeclampsia (PE) with concomitant intrauterine growth restriction (IUGR). TNFR1 and TNFR2 were evaluated in a cross-sectional study comparing preeclamptic (n = 15) with or without IUGR versus normotensive pregnant women (PREG, n = 30), and non-pregnant controls (Con; n = 20). Plasma levels of sTNFR1 were higher in PE (1675.0 ± 227.1 pg/mL) compared with PREG (1035.0 ± 101.1 pg/mL) and Con (589.3 ± 82.67 pg/mL), with the highest values observed in PE with IUGR (2624.0 ± 421.4 pg/mL; n = 6). Plasma sTNFR2 was higher during pregnancy (PE: 1836.0 ± 198.7 pg/mL; PREG: 1697.0 ± 95.0 pg/mL) compared with Con (598.3 ± 82.7 pg/mL). Urinary levels of sTNFR1 and sTNFR2 were higher in PE and PREG compared with the Con group. Abundance of TNFR1 mRNA in peripheral blood leukocytes was strongly correlated with plasma levels of sTNFR1 in PE. However, TNFR2 mRNA accumulation in leukocytes did not correlate with sTNFR2 plasma levels. The level of sTNFR1 in plasma was correlated with body weight of the newborn (r = -0.56). The data suggest that maternal leukocytes contribute to sTNFR1 levels in plasma in association with decreasing newborn weight and PE with concomitant IUGR.


Assuntos
Retardo do Crescimento Fetal/imunologia , Leucócitos/metabolismo , Pré-Eclâmpsia/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/sangue , Receptores Tipo I de Fatores de Necrose Tumoral/sangue , Adulto , Biomarcadores/sangue , Biomarcadores/urina , Peso ao Nascer , Estudos de Casos e Controles , Estudos Transversais , Feminino , Retardo do Crescimento Fetal/sangue , Retardo do Crescimento Fetal/urina , Humanos , Recém-Nascido , Pessoa de Meia-Idade , Pré-Eclâmpsia/sangue , Pré-Eclâmpsia/urina , Gravidez , Reação em Cadeia da Polimerase em Tempo Real , Receptores Tipo I de Fatores de Necrose Tumoral/urina , Receptores Tipo II do Fator de Necrose Tumoral/urina , Índice de Gravidade de Doença
10.
Hypertension ; 63(3): 551-8, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24324041

RESUMO

A recent genome-wide association study identified a locus on chromosome 16 in the promoter region of the uromodulin (UMOD) gene that is associated with hypertension. Here, we examined the hypertension signal with functional studies in Umod knockout (KO) mice. Systolic blood pressure was significantly lower in KO versus wild-type (WT) mice under basal conditions (KO: 116.6±0.3 mm Hg versus WT: 136.2±0.4 mm Hg; P<0.0001). Administration of 2% NaCl did not alter systolic blood pressure in KO mice, whereas it increased in WT mice by ≈33%, P<0.001. The average 24-hour urinary sodium excretion in the KO was greater than that of WT mice (P<0.001). Chronic renal function curves demonstrate a leftward shift in KO mice, suggesting that the relationship between UMOD and blood pressure is affected by sodium. Creatinine clearance was increased during salt loading with 2% NaCl in the KO mice, leading to augmented filtered Na(+) excretion and further Na(+) loss. The difference in sodium uptake that exists between WT and KO strains was explored at the molecular level. Urinary tumor necrosis factor-α levels were significantly higher in KO mice compared with WT mice (P<0.0001). Stimulation of primary thick ascending limb of the loop of Henle cells with exogenous tumor necrosis factor-α caused a reduction in NKCC2A expression (P<0.001) with a concurrent rise in the levels of UMOD mRNA (P<0.001). Collectively, we demonstrate that UMOD regulates sodium uptake in the thick ascending limb of the loop of Henle by modulating the effect of tumor necrosis factor-α on NKCC2A expression, making UMOD an important determinant of blood pressure control.


Assuntos
Pressão Sanguínea/fisiologia , Regulação da Expressão Gênica , Hipertensão/genética , RNA/genética , Uromodulina/genética , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Hipertensão Essencial , Humanos , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/metabolismo , Uromodulina/biossíntese
11.
Free Radic Biol Med ; 63: 108-14, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23619127

RESUMO

Mitochondrial diseases (MDs) are heterogeneous disorders due to impaired respiratory chain function causing defective ATP production. Although the disruption of oxidative phosphorylation is central to the MD pathophysiology, other factors may contribute to these disorders. We investigated the expression and the cellular localization of TNF-α and its receptors, TNFR1 and TNFR2, in muscle biopsies from 15 patients with mitochondrial respiratory chain dysfunction. Our data unambiguously demonstrate that TNF-α is expressed in muscle fibers with abnormal focal accumulations of mitochondria, so-called ragged red fibers, and is delivered to mitochondria where both receptors are localized. Moreover TNF receptors are differentially regulated in patients' muscle in which the expression levels of TNFR1 mRNA are decreased and those of TNFR2 mRNA are increased compared with controls. These findings suggest for the first time that TNF-α could exert a direct effect on mitochondria via its receptors.


Assuntos
Doenças Mitocondriais/metabolismo , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Biópsia , DNA Mitocondrial/genética , Feminino , Expressão Gênica , Humanos , Masculino , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Mutação , Fosforilação Oxidativa , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Receptores Tipo II do Fator de Necrose Tumoral/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/genética
12.
Am J Physiol Renal Physiol ; 304(5): F533-42, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23269645

RESUMO

Pathways that contribute to TNF production by the kidney are not well defined. Mice given 1% NaCl in the drinking water for 3 days exhibited a 2.5-fold increase in urinary, but not plasma, TNF levels compared with mice given tap water. Since furosemide attenuated the increase in TNF levels, we hypothesized that hypertonic NaCl intake increases renal TNF production by a pathway involving the Na(+)-K(+)-2Cl(-) cotransporter (NKCC2). A 2.5-fold increase in NKCC2A mRNA accumulation was observed in medullary thick ascending limb (mTAL) tubules from mice given 1% NaCl; a concomitant 2-fold increase in nuclear factor of activated T cells 5 (NFAT5) mRNA and protein expression was observed in the outer medulla. Urinary TNF levels were reduced in mice given 1% NaCl after an intrarenal injection of a lentivirus construct designed to specifically knockdown NKCC2A (EGFP-N2A-ex4); plasma levels of TNF did not change after injection of EGFP-N2A-ex4. Intrarenal injection of EGFP-N2A-ex4 also inhibited the increase of NFAT5 mRNA abundance in the outer medulla of mice given 1% NaCl. TNF production by primary cultures of mTAL cells increased approximately sixfold in response to an increase in osmolality to 400 mosmol/kgH2O produced with NaCl and was inhibited in cells transiently transfected with a dnNFAT5 construct. Transduction of cells with EGFP-N2A-ex4 also prevented increases in TNF mRNA and protein production in response to high NaCl concentration and reduced transcriptional activity of a NFAT5 promoter construct. Since NKCC2A expression is restricted to the TAL, NKCC2A-dependent activation of NFAT5 is part of a pathway by which the TAL produces TNF in response to hypertonic NaCl intake.


Assuntos
Rim/metabolismo , Fatores de Transcrição NFATC/metabolismo , Cloreto de Sódio/farmacologia , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células Cultivadas , Rim/citologia , Rim/efeitos dos fármacos , Alça do Néfron/citologia , Alça do Néfron/efeitos dos fármacos , Alça do Néfron/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 1 da Família 12 de Carreador de Soluto , Fator de Necrose Tumoral alfa/urina
13.
Prostaglandins Other Lipid Mediat ; 99(1-2): 45-50, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22800939

RESUMO

The effect of tumor necrosis factor-alpha (TNF) on cyclooxygenase-2 (COX-2) expression in the renal outer medulla (OM) was determined in a model of dihydrotachysterol (DHT)-induced hypercalcemia. Increases in serum calcium and water intake were observed during ingestion of a DHT-containing diet in both wild type (WT) and TNF deficient mice (TNF(-/-)). Polyuria and a decrease in body weight were observed in response to DHT treatment in WT and TNF(-/-) mice. A transient elevation in urinary TNF was observed in WT mice treated with DHT. Moreover, increased urinary levels of prostaglandin E(2) (PGE(2)) and a corresponding increase in COX-2 expression in the OM were observed in WT mice fed DHT. Increased COX-2 expression was not observed in TNF(-/-) mice fed DHT, and the characteristics of PGE(2) synthesis were distinct from those in WT mice. This study demonstrates that COX-2 expression in the OM, secondary to hypercalemia, is TNF-dependent.


Assuntos
Ciclo-Oxigenase 2/biossíntese , Hipercalcemia/metabolismo , Medula Renal/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Di-Hidrotaquisterol , Hipercalcemia/induzido quimicamente , Masculino , Camundongos , Poliúria/induzido quimicamente , Fator de Necrose Tumoral alfa/deficiência , Fator de Necrose Tumoral alfa/urina
14.
Prostaglandins Other Lipid Mediat ; 98(3-4): 101-6, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22101002

RESUMO

The thick ascending limb of Henle's loop (TAL) is capable of metabolizing arachidonic acid (AA) by cytochrome P450 (CYP450) and cyclooxygenase (COX) pathways and has been identified as a nephron segment that contributes to salt-sensitive hypertension. Previous studies demonstrated a prominent role for CYP450-dependent metabolism of AA to products that inhibited ion transport pathways in the TAL. However, COX-2 is constitutively expressed along all segments of the TAL and is increased in response to diverse stimuli. The ability of Tamm-Horsfall glycoprotein, a selective marker of cortical TAL (cTAL) and medullary (mTAL), to bind TNF and localize it to this nephron segment prompted studies to determine the capacity of mTAL cells to produce TNF and determine its effects on mTAL function. The colocalization of calcium-sensing receptor (CaR) and COX-2 in the TAL supports the notion that activation of CaR induces TNF-dependent COX-2 expression and PGE2 synthesis in mTAL cells. Additional studies showed that TNF produced by mTAL cells inhibits 86Rb uptake, an in vitro correlate of natriuresis, in an autocrine- and COX-2-dependent manner. The molecular mechanism for these effects likely includes inhibition of Na⁺-K⁺-2Cl⁻ cotransporter (NKCC2) expression and trafficking.


Assuntos
Eicosanoides/metabolismo , Alça do Néfron/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Humanos , Alça do Néfron/enzimologia , Receptores de Detecção de Cálcio/metabolismo
15.
Am J Physiol Renal Physiol ; 301(1): F94-100, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21511694

RESUMO

The effects of TNF gene deletion on renal Na(+)-K(+)-2Cl(-) cotransporter (NKCC2) expression and activity were determined. Outer medulla from TNF(-/-) mice exhibited a twofold increase in total NKCC2 protein expression compared with wild-type (WT) mice. This increase was not observed in TNF(-/-) mice treated with recombinant human TNF (hTNF) for 7 days. Administration of hTNF had no effect on total NKCC2 expression in WT mice. A fourfold increase in NKCC2A mRNA accumulation was observed in outer medulla from TNF(-/-) compared with WT mice; NKCC2F and NKCC2B mRNA accumulation was similar between genotypes. The increase in NKCC2A mRNA accumulation was attenuated when TNF(-/-) mice were treated with hTNF. Bumetanide-sensitive O(2) consumption, an in vitro correlate of NKCC2 activity, was 2.8 ± 0.2 nmol·min(-1)·mg(-1) in medullary thick ascending limb tubules from WT, representing ∼40% of total O(2) consumption, whereas, in medullary thick ascending limb tubules from TNF(-/-) mice, it was 5.6 ± 0.3 nmol·min(-1)·mg(-1), representing ∼60% of total O(2) consumption. Administration of hTNF to TNF(-/-) mice restored the bumetanide-sensitive component to ∼30% of total O(2) consumption. Ambient urine osmolality was higher in TNF(-/-) compared with WT mice (2,072 ± 104 vs. 1,696 ± 153 mosmol/kgH(2)O, P < 0.05). The diluting ability of the kidney, assessed by measuring urine osmolality before and after 1 h of water loading also was greater in TNF(-/-) compared with WT mice (174 ± 38 and 465 ± 81 mosmol/kgH(2)O, respectively, P < 0.01). Collectively, these findings suggest that TNF plays a role as an endogenous inhibitor of NKCC2 expression and function.


Assuntos
Medula Renal/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Fator de Necrose Tumoral alfa/fisiologia , Animais , Western Blotting , Cloretos/metabolismo , Fragmentação do DNA , DNA Complementar/biossíntese , Indicadores e Reagentes , Isomerismo , Capacidade de Concentração Renal/fisiologia , Testes de Função Renal , Medula Renal/citologia , Medula Renal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Concentração Osmolar , Consumo de Oxigênio/efeitos dos fármacos , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 1 da Família 12 de Carreador de Soluto , Fator de Necrose Tumoral alfa/genética
16.
Am J Physiol Renal Physiol ; 299(5): F1141-50, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20739394

RESUMO

The hypothesis that TNF receptor 1-deficient (TNFR1(-/-)) mice display blood pressure (BP) and renal functional responses that differ from wild-type (WT) mice was tested in an angiotensin II (ANG II)-dependent model of hypertension. Basal systolic BP (SBP), mean arterial pressure, diastolic BP, heart rate (HR), and pulse pressure were similar in WT and TNFR1(-/-) mice. Infusion of ANG II for 7 days elevated SBP to a greater extent in TNFR1(-/-) compared with WT mice; pulse pressure was also elevated in TNFR1(-/-). HR decreased in TNFR1(-/-) mice infused with ANG II, an effect prominent on day 1. Basal urinary albumin excretion was similar in WT and TNFR1(-/-) mice but was higher in TNFR1(-/-) in response to ANG II infusion. Water intake and urine volume were increased by ANG II infusion; this increase was higher in TNFR1(-/-) vs. WT mice, whereas body weight and food intake were unaffected. Baseline creatinine clearance (Ccr), urinary sodium excretion, and fractional excretion of sodium (FE(Na)%) were similar in vehicle-treated WT and TNFR1(-/-) mice. ANG II infusion for 7 days increased Ccr and filtered load of sodium in TNFR1(-/-) but not WT mice, whereas it elicited an increase in FE(Na)% and urinary sodium excretion in WT but not TNFR1(-/-) mice. ANG II also inhibited renal TNFR1 mRNA accumulation while increasing that of TNFR2. These findings indicate deletion of TNFR1 is associated with an exacerbated SBP response, decrease in HR, and altered renal function in ANG II-dependent hypertension.


Assuntos
Angiotensina II/farmacologia , Pressão Sanguínea/fisiologia , Rim/efeitos dos fármacos , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/genética , Vasoconstritores/farmacologia , Animais , Peso Corporal/fisiologia , Ingestão de Alimentos/fisiologia , Frequência Cardíaca/efeitos dos fármacos , Frequência Cardíaca/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Circulação Renal/efeitos dos fármacos , Telemetria , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/genética
17.
Am J Physiol Renal Physiol ; 296(6): F1494-503, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19369291

RESUMO

The contribution of nuclear factor of activated T cells 5 (NFAT5) to the regulation of tumor necrosis factor-alpha (TNF) production in medullary thick ascending limb (mTAL) cells is unclear. RT-PCR analysis was performed on primary cultures of mouse mTAL cells and freshly isolated mTAL tubules to determine which NFAT isoforms are present in this nephron segment. Primer pairs were designed, based on published sequences for mouse NFAT1-5, to produce fragments of approximately 200 bp. Analysis of PCR products by gel electrophoresis and subsequent DNA sequencing indicated that cells and tubules contained mRNA for all five NFAT isoforms. The relative expression of NFAT isoforms was then determined using quantitative real-time RT-PCR. The data indicate that NFAT isoforms 5 >/= 1 are the predominant isoforms present in mTAL cells and tubules. Western blot analysis demonstrated constitutive expression of NFAT5 in nuclear extracts from mTAL tubules and primary culture cells; expression in mTAL cells also was detected by immunofluorescence. Expression of NFAT5 was increased in mTAL cells transiently transfected with an NFAT5 overexpression vector (pcDNA3.1-NFAT5), resulting in increased basal and calcium-sensing receptor (CaR)-mediated TNF production. Transient transfection of mTAL cells with a small hairpin RNA vector that targeted exon 8 of NFAT5 (U6-N5 ex8) significantly inhibited TNF promoter activity. Transient transfection with U6-N5 ex8 also reduced nuclear expression of NFAT5, TNF mRNA accumulation, and attenuated CaR-mediated activation of Cl(-) entry into polarized mTAL cells. Collectively, these data suggest that activation of NFAT5 is part of a TNF-dependent pathway that inhibits apical Cl(-) influx in the mTAL after activation of CaR.


Assuntos
Regulação da Expressão Gênica/fisiologia , Alça do Néfron/citologia , Alça do Néfron/metabolismo , Fatores de Transcrição/metabolismo , Animais , Células Cultivadas , Ciclo-Oxigenase 2/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transporte Proteico , RNA Mensageiro/metabolismo , Receptores de Detecção de Cálcio/metabolismo , Cloreto de Sódio/metabolismo , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo
18.
Am J Physiol Renal Physiol ; 293(4): F1413-22, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17670898

RESUMO

A medullary thick ascending limb (mTAL) cell line, termed raTAL, has been established from freshly isolated rat mTAL tubules and cultured continuously for up to 75 passages; it retains characteristics of mTAL cells even after retrieval from storage in liquid nitrogen for several months. The cells express Tamm-Horsfall glycoprotein (THP), a TAL-specific marker, grow to confluence, exhibit a polygonal morphology characteristic of epithelial cells, and form "domes." Detection of THP, Na(+)-K(+)-2Cl(-) cotransporter (NKCC2), Na(+)-K(+)-ATPase, and renal outer medullary K(+) channel (ROMK) was achieved using indirect immunofluorescence and confocal microscopy. Western blot analysis of NKCC2 expression using two different antibodies revealed a band of approximately 160 kDa, and RT-PCR analysis demonstrated the presence of NKCC2 isoforms A and F, which was confirmed by DNA sequencing; transport of Cl(-) into raTAL cells was inhibited by furosemide. Ouabain- and bumetanide-sensitive oxygen consumption, an index of ion transport activity in the mTAL, was observed in raTAL cells, and the number of domes present was reduced significantly when cells were incubated in the presence of ouabain or bumetanide. The specific activity of Na(+)-K(+)-ATPase activity was determined in raTAL cells (0.67 +/- 0.18 nmol P(i).microg protein(-1).min(-1)), primary cultures of mTAL cells (0.39 +/- 0.08 nmol P(i).microg protein(-1).min(-1)), and freshly isolated mTAL tubules (1.10 +/- 0.29 nmol P(i).microg protein(-1).min(-1)), and approximately 30-50% of total cellular ATPase activity was inhibited by ouabain, in accord with other mTAL preparations. This cell line will be used in studies that address biochemical, molecular, and physiological mechanisms in the mTAL.


Assuntos
Linhagem Celular/fisiologia , Medula Renal/citologia , Medula Renal/metabolismo , Alça do Néfron/citologia , Alça do Néfron/metabolismo , Animais , Cloretos/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Masculino , Mucoproteínas/metabolismo , Consumo de Oxigênio/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Ratos , Ratos Sprague-Dawley , Simportadores de Cloreto de Sódio-Potássio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Membro 1 da Família 12 de Carreador de Soluto , Uromodulina
19.
Am J Physiol Renal Physiol ; 290(5): F1110-7, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16380462

RESUMO

Because nuclear factor of activated T cells (NFAT) has been implicated in TNF production as well as osmoregulation and salt and water homeostasis, we addressed whether calcium-sensing receptor (CaR)-mediated TNF production in medullary thick ascending limb (mTAL) cells was NFAT dependent. TNF production in response to addition of extracellular Ca(2+) (1.2 mM) was abolished in mTAL cells transiently transfected with a dominant-negative CaR construct (R796W) or pretreated with the phosphatidylinositol phospholipase C (PI-PLC) inhibitor U-73122. Cyclosporine A (CsA), an inhibitor of the serine/threonine phosphatase calcineurin, and a peptide ligand, VIVIT, that selectively inhibits calcineurin-NFAT signaling, also prevented CaR-mediated TNF production. Increases in calcineurin activity in cells challenged with Ca(2+) were inhibited after pretreatment with U-73122 and CsA, suggesting that CaR activation increases calcineurin activity in a PI-PLC-dependent manner. Moreover, U-73122, CsA, and VIVIT inhibited CaR-dependent activity of an NFAT construct that drives expression of firefly luciferase in transiently transfected mTAL cells. Collectively, these data verify the role of calcineurin and NFAT in CaR-mediated TNF production by mTAL cells. Activation of the CaR also increased the binding of NFAT to a consensus oligonucleotide, an effect that was blocked by U-73122 and CsA, suggesting that a calcineurin- and NFAT-dependent pathway increases TNF production in mTAL cells. This mechanism likely regulates TNF gene transcription as U-73122, CsA, and VIVIT blocked CaR-dependent activity of a TNF promoter construct. Elucidating CaR-mediated signaling pathways that regulate TNF production in the mTAL will be crucial to understanding mechanisms that regulate extracellular fluid volume and salt balance.


Assuntos
Fatores de Transcrição NFATC/fisiologia , Receptores de Detecção de Cálcio/fisiologia , Fator de Necrose Tumoral alfa/biossíntese , Animais , Calcineurina/fisiologia , Técnicas de Cultura de Células , Medula Renal/citologia , Medula Renal/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Transcrição Gênica , Transfecção
20.
Wei Sheng Yan Jiu ; 31(4): 229-31, 2002 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-12600024

RESUMO

Metallothionein (MT), a low-molecular-weight, cysteine-rich, zinc-binding protein, has an important effect in the detoxification of various metals. However, extra Zn7-MT protects aginst the cellular toxicity of lead in rat primary hepatocyte (RPHC) culture has seldom been examined thoroughly. This study was therefore designed to determine the effects of extra Zn7-MT on the toxicity of lead in rat primary hepatocyte culture. Hepatocytes were grown in monolayer culture for 24 h and Zn7-MT was subsequently added for 24 h. RPHC were exposed to different concentration of lead for 24 h. Cytotoxicity was assessed by enzyme leakage and loss of intracellular K+. Meanwhile, the subcellular distribution and accumulation of lead was studied, and the protactive effect on the cellular toxicity of lead in RPHC culture in the present of Zn7-MT was investigated. The results showed that the toxicity of lead was significantly less in the presence of Zn7-MT. The hepatocelluar uptake and accumulation of lead have markedly altered. In the cytosol of control cells, the lead was bound mainly to high-molecular-weight proteins whereas the lead was mainly associated with MT in the presence of extra Zn7-MT.


Assuntos
Hepatócitos/citologia , Chumbo/toxicidade , Metalotioneína/farmacologia , Zinco/farmacologia , Animais , Células Cultivadas , Masculino , Coelhos , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA