Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Pregnancy Hypertens ; 2(3): 208, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-26105272

RESUMO

INTRODUCTION: In pregnancy, plasma volume is expanded due to high aldosterone levels to support placental perfusion and fetal nutrition. Inadequately low aldosterone levels as present in preeclampsia, a life-threatening disease for both mother and child, are discussed to be involved in its pathogenesis or severity. OBJECTIVES: We used aldosterone synthase deficient (AS(-/-)) mice to test whether the absence of aldosterone is sufficient to impair pregnancy or even to cause preeclampsia. METHODS: AS(-/-) and AS(+/+) females were mated with AS(+/+) and AS(-/-) males, respectively, always generating AS(+/-) offspring. Blood pressure was measured by tail cuff, fetal and placental number and size as well as placental histology were assessed. Placental expression of HIF-1αand angiogenic factors was assessed by semiquantitative RT-PCR. RESULTS: With maternal aldosterone deficiency in AS(-/-) mice, systolic blood pressure was low before and further reduced during pregnancy and with no increase in proteinuria. Yet, AS(-/-) had smaller litters due to loss of fetuses as indicated by a high number of necrotic placentas with massive lymphocyte infiltrations at gestational day 18. Surviving fetuses and their placentas from AS(-/-) females were smaller. High salt diet before and during pregnancy increased systolic blood pressure only before pregnancy in both genotypes and reduced blood pressure during late pregnancy as compared to normal salt controls. Litter size from AS(-/-) was slightly improved and the differences in placental and fetal weights between AS(+/+) and AS(-/-) mothers disappeared. Overall an increased placental efficiency was observed in both groups. CONCLUSION: Our results demonstrate that aldosterone deficiency has profound adverse effects on placental function. High dietary salt intake improved placental function and lowered blood pressure in wild-type mice. In this animal model, aldosterone deficiency did not cause preeclampsia.

2.
Kidney Int ; 70(10): 1706-16, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16985514

RESUMO

Final urinary acidification is achieved by electrogenic vacuolar H(+)-ATPases expressed in acid-secretory intercalated cells (ICs) in the connecting tubule (CNT) and the cortical (CCD) and initial medullary collecting duct (MCD), respectively. Electrogenic Na(+) reabsorption via epithelial Na(+) channels (ENaCs) in the apical membrane of the segment-specific CNT and collecting duct cells may promote H(+)-ATPases-mediated proton secretion by creating a more lumen-negative voltage. The exact localization where this supposed functional interaction takes place is unknown. We used several mouse models performing renal clearance experiments and assessed the furosemide-induced urinary acidification. Increasing Na(+) delivery to the CNT and CCD by blocking Na(+) reabsorption in the thick ascending limb with furosemide enhanced urinary acidification and net acid excretion. This effect of furosemide was abolished with amiloride or benzamil blocking ENaC action. In mice deficient for the IC-specific B1 subunit of the vacuolar H(+)-ATPase, furosemide led to only a small urinary acidification. In contrast, in mice with a kidney-specific inactivation of the alpha subunit of ENaC in the CCD and MCD, but not in the CNT, furosemide alone and in combination with hydrochlorothiazide induced normal urinary acidification. These results suggest that the B1 vacuolar H(+)-ATPase subunit is necessary for the furosemide-induced acute urinary acidification. Loss of ENaC channels in the CCD and MCD does not affect this acidification. Thus, functional expression of ENaC channels in the CNT is sufficient for furosemide-stimulated urinary acidification and identifies the CNT as a major segment in electrogenic urinary acidification.


Assuntos
Equilíbrio Ácido-Base/efeitos dos fármacos , Diuréticos/farmacologia , Furosemida/farmacologia , Túbulos Renais Distais/efeitos dos fármacos , ATPases Translocadoras de Prótons/metabolismo , Equilíbrio Ácido-Base/fisiologia , Amilorida/farmacocinética , Amilorida/farmacologia , Animais , Diuréticos/farmacocinética , Canais Epiteliais de Sódio/genética , Canais Epiteliais de Sódio/metabolismo , Furosemida/farmacocinética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Enzimológica da Expressão Gênica/genética , Hidroclorotiazida/farmacocinética , Hidroclorotiazida/farmacologia , Concentração de Íons de Hidrogênio , Túbulos Renais Coletores/metabolismo , Túbulos Renais Distais/metabolismo , Taxa de Depuração Metabólica/efeitos dos fármacos , Taxa de Depuração Metabólica/fisiologia , Camundongos , Camundongos Knockout , Néfrons/efeitos dos fármacos , Néfrons/fisiologia , ATPases Translocadoras de Prótons/genética , Equilíbrio Hidroeletrolítico/fisiologia
3.
Kidney Int ; 69(6): 1024-32, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16528252

RESUMO

Previous studies revealed that chronic (days) vasopressin treatment stimulates amiloride-sensitive sodium transport in isolated renal cortical collecting ducts and increases the abundance of beta- and gamma-subunits of the epithelial sodium channel (ENaC) in the kidney. The aim of the present work was to investigate in vivo the cellular basis of these effects. The long-term effect of V2 vasopressin agonist (1-deamino-8-D-arginine vasopressin (dDAVP)) on the abundance and subcellular localization of ENaC along the rat renal collecting system was determined by immunohistochemistry and laser confocal microscopy. Moreover, we studied by real-time reverse transcriptase-polymerase chain reaction the effect of vasopressin on proteins implicated in the regulation of ENaC (Nedd4-2, prostasin, Sgk1). After 5 days of administration, dDAVP markedly increased the intracellular pool of the beta- and gamma-ENaC subunits in the principal cells, with an increasing gradient from connecting tubule to the outer medullary collecting duct, but did not increase any subunit at the cell surface. The apical immunostaining of ENaC increased in response to sodium restriction, as expected, but dDAVP did not further enhance this apical labelling. dDAVP increased the gene expression of prostasin in the cortex but not that of Nedd4-2 and Sgk1. These findings suggest that the previously reported increase in sodium transport induced by sustained stimulation of vasopressin V2 receptor is probably mediated by other mechanism than an increase in the apical density of ENaC.


Assuntos
Túbulos Renais Coletores/química , Canais de Sódio/análise , Vasopressinas/farmacologia , Aldosterona/análise , Aldosterona/sangue , Animais , Desamino Arginina Vasopressina/farmacologia , Ingestão de Líquidos , Canais Epiteliais de Sódio , Imunofluorescência , Expressão Gênica/efeitos dos fármacos , Imuno-Histoquímica , Córtex Renal/química , Masculino , Microscopia Confocal , Ratos , Ratos Brattleboro , Ratos Sprague-Dawley , Receptores de Vasopressinas/análise , Receptores de Vasopressinas/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serina Endopeptidases/análise , Serina Endopeptidases/genética , Sódio na Dieta/farmacologia , Fatores de Tempo , Vasopressinas/sangue
4.
EMBO J ; 20(24): 7052-9, 2001 Dec 17.
Artigo em Inglês | MEDLINE | ID: mdl-11742982

RESUMO

The epithelial Na(+) channel (ENaC) plays an essential role in the regulation of whole body Na(+) balance and blood pressure. The cell surface expression of this channel, a complex of three subunits (alpha, beta and gamma ENaC), has been shown to be regulated by hormones such as aldosterone and vasopressin and by intracellular signaling, including ubiquitylation and/or phosphorylation. However, the molecular mechanisms involving phosphorylation in the regulation of ENaC are unclear. Here we show by expression studies in Xenopus laevis oocytes that the aldosterone-induced Sgk1 kinase interacts with the ubiquitin protein ligase Nedd4-2 in a PY motif-dependent manner and phosphorylates Nedd4-2 on Ser444 and, to a lesser extent, Ser338. Such phosphorylation reduces the interaction between Nedd4-2 and ENaC, leading to elevated ENaC cell surface expression. These data show that phosphorylation of an enzyme involved in the ubiquitylation cascade (Nedd4-2) controls cell surface density of ENaC and propose a paradigm for the control of ion channels. Moreover, they suggest a novel and complete signaling cascade for aldosterone-dependent regulation of ENaC.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Ligases/metabolismo , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Sódio/metabolismo , Ubiquitina-Proteína Ligases , Sequência de Aminoácidos , Animais , Linhagem Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte , Canais Epiteliais de Sódio , Proteínas Imediatamente Precoces , Ubiquitina-Proteína Ligases Nedd4 , Oócitos/metabolismo , Fosforilação , Ligação Proteica , Ubiquitina/metabolismo , Proteínas de Xenopus , Xenopus laevis
5.
Am J Physiol Renal Physiol ; 281(6): F1021-7, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11704552

RESUMO

The organization of Na(+) and Ca(2+) transport pathways along the mouse distal nephron is incompletely known. We revealed by immunohistochemistry a set of Ca(2+) and Na(+) transport proteins along the mouse distal convolution. The thiazide-sensitive Na(+)-Cl(-) cotransporter (NCC) characterized the distal convoluted tubule (DCT). The amiloride-sensitive epithelial Na(+) channel (ENaC) colocalized with NCC in late DCT (DCT2) and extended to the downstream connecting tubule (CNT) and collecting duct (CD). In early DCT (DCT1), the basolateral Ca(2+)-extruding proteins [Na(+)/Ca(2+) exchanger (NCX), plasma membrane Ca(2+)-ATPase (PCMA)] and the cytoplasmic Ca(2+)-binding protein calbindin D(28K) (CB) were found at very low levels, whereas the cytoplasmic Ca(2+)/Mg(2+)-binding protein parvalbumin was highly abundant. NCX, PMCA, and CB prevailed in DCT2 and CNT, where we located the apical epithelial Ca(2+) channel (ECaC1). Its subcellular localization changed from apical in DCT2 to exclusively cytoplasmic at the end of CNT. NCX and PMCA decreased in parallel with the fading of ECaC1 in the apical membrane. All three of them were undetectable in CD. These findings disclose DCT2 and CNT as major sites for transcellular Ca(2+) transport in the mouse distal nephron. Cellular colocalization of Ca(2+) and Na(+) transport pathways suggests their mutual interactions in transport regulation.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte/análise , Túbulos Renais Distais/metabolismo , Receptores de Droga , Sódio/metabolismo , Simportadores , Animais , Calbindinas , Canais de Cálcio/análise , Canais de Cálcio/imunologia , ATPases Transportadoras de Cálcio/análise , ATPases Transportadoras de Cálcio/imunologia , Proteínas de Transporte/imunologia , Proteínas de Transporte de Cátions , Canais Epiteliais de Sódio , Feminino , Imuno-Histoquímica , Transporte de Íons , Túbulos Renais Distais/química , Camundongos , Modelos Biológicos , Parvalbuminas/análise , Parvalbuminas/imunologia , ATPases Transportadoras de Cálcio da Membrana Plasmática , Proteína G de Ligação ao Cálcio S100/análise , Proteína G de Ligação ao Cálcio S100/imunologia , Canais de Sódio/análise , Canais de Sódio/imunologia , Simportadores de Cloreto de Sódio , Trocador de Sódio e Cálcio/análise , Trocador de Sódio e Cálcio/imunologia , Membro 3 da Família 12 de Carreador de Soluto , Canais de Cátion TRPV
6.
Am J Physiol Cell Physiol ; 281(6): C1889-97, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11698247

RESUMO

The DeltaF508 mutation reduces the amount of cystic fibrosis transmembrane conductance regulator (CFTR) expressed in the plasma membrane of epithelial cells. However, a reduced temperature, butyrate compounds, and "chemical chaperones" allow DeltaF508-CFTR to traffic to the plasma membrane and increase Cl(-) permeability in heterologous and nonpolarized cells. Because trafficking is affected by the polarized state of epithelial cells and is cell-type dependent, our goal was to determine whether these maneuvers induce DeltaF508-CFTR trafficking to the apical plasma membrane in polarized epithelial cells. To this end, we generated and characterized a line of polarized Madin-Darby canine kidney (MDCK) cells stably expressing DeltaF508-CFTR tagged with green fluorescent protein (GFP). A reduced temperature, glycerol, butyrate, or DMSO had no effect on 8-(4-chlorophenylthio)-cAMP (CPT-cAMP)-stimulated transepithelial Cl(-) secretion across polarized monolayers. However, when the basolateral membrane was permeabilized, butyrate, but not the other experimental maneuvers, increased the CPT-cAMP-stimulated Cl(-) current across the apical plasma membrane. Thus butyrate increased the amount of functional DeltaF508-CFTR in the apical plasma membrane. Butyrate failed to stimulate transepithelial Cl(-) secretion because of inhibitory effects on Cl(-) uptake across the basolateral membrane. These observations suggest that studies on heterologous and nonpolarized cells should be interpreted cautiously. The GFP tag on DeltaF508-CFTR will allow investigation of DeltaF508-CFTR trafficking in living, polarized MDCK epithelial cells in real time.


Assuntos
Membrana Celular/metabolismo , Polaridade Celular , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Proteínas Luminescentes/metabolismo , Animais , Butiratos/farmacologia , Bloqueadores dos Canais de Cálcio/farmacologia , Linhagem Celular , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Genisteína/farmacologia , Proteínas de Fluorescência Verde , Humanos , Indicadores e Reagentes/metabolismo , Ionóforos/farmacologia , Proteínas Luminescentes/genética , Microscopia Confocal , Nistatina/farmacologia , Transporte Proteico , Proteínas Recombinantes de Fusão/metabolismo , Temperatura , Transgenes , ortoaminobenzoatos/farmacologia
7.
Curr Opin Nephrol Hypertens ; 10(5): 667-75, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11496063

RESUMO

The aldosterone-sensitive distal nephron extends from the second part of the distal convoluted tubule to the inner medullary collecting duct. As recently shown, aldosterone increases within two hours the abundance of the alpha-subunit of the epithelial sodium channel along the entire aldosterone-sensitive distal nephron, whereas it induces only in an initial portion of the aldosterone-sensitive distal nephron an apical translocation of all three epithelial sodium channel subunits. This suggests that another factor or factors determines the length of the aldosterone-sensitive distal nephron portion in which aldosterone controls epithelial sodium channel surface expression. Since the glucocorticoid-induced kinase SGK1 was identified as aldosterone-induced protein in 1999, it has been postulated to play a key regulatory role. The in-vivo localization of its induction to segment-specific cells of the aldosterone-sensitive distal nephron, and the in-vitro correlation of the amount of its hyperphosphorylated form with transepithelial sodium transport, support this hypothesis. Other recent studies unravel pathways other than those activated by aldosterone and insulin that impact on SGK1 expression and/or function, and thus shed some light onto the complex network that appears to control sodium transport. In view of the ongoing research, the question of how, and formally also whether, SGK1 acts on the epithelial sodium channel should be resolved in the near future.


Assuntos
Aldosterona/fisiologia , Túbulos Renais Distais/fisiologia , Proteínas Nucleares , Animais , Canais Epiteliais de Sódio , Proteínas Imediatamente Precoces , Proteínas Serina-Treonina Quinases/fisiologia , Canais de Sódio/metabolismo
8.
Am J Physiol Renal Physiol ; 280(4): F675-82, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11249859

RESUMO

Aldosterone controls sodium reabsorption and potassium secretion in the aldosterone-sensitive distal nephron (ASDN). Although clearance measurements have shown that aldosterone induces these transports within 30--60 min, no early effects have been demonstrated in vivo at the level of the apical epithelial sodium channel (ENaC), the main effector of this regulation. Here we show by real-time RT-PCR and immunofluorescence that an aldosterone injection in adrenalectomized rats induces alpha-ENaC subunit expression along the entire ASDN within 2 h, whereas beta- and gamma-ENaC are constitutively expressed. In the proximal ASDN portions only, ENaC is shifted toward the apical cellular pole and the apical plasma membrane within 2 and 4 h, respectively. To address the question of whether the early aldosterone-induced serum and glucocorticoid-regulated kinase (SGK) might mediate this apical shift of ENaC, we analyzed SGK induction in vivo. Two hours after aldosterone, SGK was highly induced in all segment-specific cells of the ASDN, and its level decreased thereafter. In Xenopus laevis oocytes, SGK induced ENaC activation and surface expression by a kinase activity-dependent mechanism. In conclusion, the rapid in vivo accumulation of SGK and alpha-ENaC after aldosterone injection takes place along the entire ASDN, whereas the translocation of alpha,beta,gamma-ENaC to the apical plasma membrane is restricted to its proximal portions. Results from oocyte experiments suggest the hypothesis that a localized activation of SGK may play a role in the mediation of ENaC translocation.


Assuntos
Aldosterona/farmacologia , Túbulos Renais Coletores/enzimologia , Proteínas Nucleares , Proteínas Serina-Treonina Quinases/metabolismo , Canais de Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Canais Epiteliais de Sódio , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Proteínas Imediatamente Precoces , Técnicas In Vitro , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Oócitos/fisiologia , Técnicas de Patch-Clamp , Proteínas Serina-Treonina Quinases/genética , RNA Mensageiro/análise , Ratos , Ratos Wistar , Sódio/metabolismo , Canais de Sódio/genética , Xenopus laevis
9.
Am J Physiol Cell Physiol ; 279(4): C1016-23, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11003582

RESUMO

Serous cells secrete Cl(-) and HCO(3)(-) and play an important role in airway function. Recent studies suggest that a Cl(-)/HCO(3)(-) anion exchanger (AE) may contribute to Cl(-) secretion by airway epithelial cells. However, the molecular identity, the cellular location, and the contribution of AEs to Cl(-) secretion in serous epithelial cells in tracheal submucosal glands are unknown. The goal of the present study was to determine the molecular identity, the cellular location, and the role of AEs in the function of serous epithelial cells. To this end, Calu-3 cells, a human airway cell line with a serous-cell phenotype, were studied by RT-PCR, immunoblot, and electrophysiological analysis to examine the role of AEs in Cl(-) secretion. In addition, the subcellular location of AE proteins was examined by immunofluorescence microscopy. Calu-3 cells expressed mRNA and protein for AE2 as determined by RT-PCR and Western blot analysis, respectively. Immunofluorescence microscopy identified AE2 in the basolateral membrane of Calu-3 cells in culture and rat tracheal serous cells in situ. In Cl(-)/HCO(3)(-)/Na(+)-containing media, the 8-(4-chlorophenylthio)adenosine 3',5'-cyclic monophosphate (CPT-cAMP)-stimulated short-circuit anion current (I(sc)) was reduced by basolateral but not by apical application of 4, 4'-diisothiocyanostilbene-2,2'-disulfonic acid (50 microM) and 4, 4'-dinitrostilbene-2,2'-disulfonic acid [DNDS (500 microM)], inhibitors of AEs. In the absence of Na(+) in the bath solutions, to eliminate the contributions of the Na(+)/HCO(3)(-) and Na(+)/K(+)/2Cl(-) cotransporters to I(sc), CPT-cAMP stimulated a small DNDS-sensitive I(sc). Taken together with previous studies, these observations suggest that a small component of cAMP-stimulated I(sc) across serous cells may be referable to Cl(-) secretion and that uptake of Cl(-) across the basolateral membrane may be mediated by AE2.


Assuntos
Proteínas de Transporte de Ânions , Antiporters/química , Antiporters/metabolismo , AMP Cíclico/análogos & derivados , Células Epiteliais/metabolismo , Mucosa Respiratória/metabolismo , Membrana Serosa/metabolismo , Animais , Antiporters/genética , Western Blotting , Linhagem Celular , Antiportadores de Cloreto-Bicarbonato , Cloretos/metabolismo , AMP Cíclico/farmacologia , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Células Epiteliais/química , Células Epiteliais/citologia , Humanos , Imuno-Histoquímica , Transporte de Íons/efeitos dos fármacos , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Isoformas de Proteínas/análise , Isoformas de Proteínas/biossíntese , RNA Mensageiro/biossíntese , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteínas SLC4A , Membrana Serosa/química , Membrana Serosa/citologia , Estilbenos/farmacologia , Tionucleotídeos/farmacologia , Traqueia
10.
Am J Physiol Renal Physiol ; 279(2): F252-8, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919843

RESUMO

Previous electrophysiological experiments on renal cortical collecting ducts indicated that dietary sodium intake and variations in aldosterone plasma levels regulate the abundance of functional epithelial Na channels (ENaC) in the apical plasma membrane. In mouse kidney we investigated by immunohistochemistry whether feeding for 3 wk a diet with high (3% Na) and low (0.05% Na) Na content influences the distribution pattern of ENaC. In mice of all experimental groups, ENaC was apparent in cells from the late portion of the distal convoluted tubule (DCT2) down to the medullary collecting duct (CD). In mice on a high-Na diet (plasma aldosterone: 40.8 +/- 2.0 ng/dl), the alpha-subunit was undetectable, and the beta- and gamma-ENaC were detected in the cytoplasm, but not in the apical plasma membrane of the cells. In contrast, in mice on a low-Na diet (plasma aldosterone: 93.6 +/- 9.3 ng/dl) all three ENaC subunits were displayed in the subapical cytoplasm and in the apical membrane of DCT2, connecting tubule (CNT), and, although less prominent, in cortical CD cells. Apical plasma membrane immunostaining progressively decreased along the cortical CD, simultaneously with increasing cytoplasmic staining for beta- and gamma-ENaC. Thus our data on mice adapted to moderately low and high Na intake suggest that regulation of ENaC function in vivo involves shifts of beta- and gamma-subunits from the cytoplasm to the apical plasma membrane and vice versa, respectively. The insertion of these subunits into the apical plasma membrane coincides with upregulation of the alpha-subunit and its insertion into the apical plasma membrane.


Assuntos
Dieta Hipossódica , Rim/metabolismo , Canais de Sódio/metabolismo , Frações Subcelulares/metabolismo , Aldosterona/sangue , Animais , Membrana Celular/metabolismo , Canais Epiteliais de Sódio , Técnicas In Vitro , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Natriurese/fisiologia , Isoformas de Proteínas/metabolismo , Sódio na Dieta/farmacologia , Distribuição Tecidual
11.
Am J Physiol Renal Physiol ; 278(4): F530-9, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10751213

RESUMO

The amiloride-sensitive epithelial sodium channel (ENaC) and the vasopressin-dependent water channel aquaporin-2 (AQP2) mediate mineralocorticoid-regulated sodium- and vasopressin-regulated water reabsorption, respectively. Distributions of ENaC and AQP2 have been shown by immunohistochemistry in rats. Functional data from rabbits suggest a different distribution pattern of these channels than in rats. We studied, by immunohistochemistry in the rabbit kidney cortex, the distributions of ENaC and AQP2, in conjunction with marker proteins for distal segments. In rabbit cortex ENaC is restricted to the connecting tubule (CNT) cells and cortical collecting duct (CCD) cells. The intracellular distribution of ENaC shifts from the apical membrane in the most upstream CNT cells to a cytoplasmic location further downstream in the CNT and in the CCD cells. AQP2 is detected in the CCD cells exclusively. The anatomic subdivisions in the rabbit distal nephron coincide exactly with distributions of apical transport systems. The differences between rabbits and rats in the distribution patterns of ENaC and AQP2 may explain functional differences in renal salt and water handling between these species.


Assuntos
Aquaporinas/metabolismo , Proteínas de Transporte/efeitos dos fármacos , Córtex Renal/metabolismo , Canais de Sódio/metabolismo , Animais , Aquaporina 2 , Aquaporina 6 , Benzotiadiazinas , Bumetanida/farmacologia , Calbindinas , Proteínas de Transporte/metabolismo , Diuréticos/farmacologia , Canais Epiteliais de Sódio , Feminino , Córtex Renal/anatomia & histologia , Masculino , ATPases Translocadoras de Prótons/metabolismo , Coelhos , Ratos , Proteína G de Ligação ao Cálcio S100/metabolismo , Inibidores de Simportadores de Cloreto de Sódio/farmacologia , Simportadores de Cloreto de Sódio-Potássio , Distribuição Tecidual
12.
Mol Biol Cell ; 10(12): 4135-47, 1999 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-10588648

RESUMO

Mutations of the glycoprotein rBAT cause cystinuria type I, an autosomal recessive failure of dibasic amino acid transport (b(0,+) type) across luminal membranes of intestine and kidney cells. Here we identify the permease-like protein b(0,+)AT as the catalytic subunit that associates by a disulfide bond with rBAT to form a hetero-oligomeric b(0,+) amino acid transporter complex. We demonstrate its b(0,+)-type amino acid transport kinetics using a heterodimeric fusion construct and show its luminal brush border localization in kidney proximal tubule. These biochemical, transport, and localization characteristics as well as the chromosomal localization on 19q support the notion that the b(0,+)AT protein is the product of the gene defective in non-type I cystinuria.


Assuntos
Sistemas de Transporte de Aminoácidos Básicos , Aminoácidos/metabolismo , Proteínas de Transporte/metabolismo , Cromossomos Humanos Par 19 , Cistinúria/metabolismo , Glicoproteínas de Membrana/metabolismo , Sequência de Aminoácidos , Sistemas de Transporte de Aminoácidos , Animais , Transporte Biológico , Proteínas de Transporte/genética , Clonagem Molecular , Cistinúria/genética , Imunofluorescência , Humanos , Hibridização In Situ , Rim/metabolismo , Rim/ultraestrutura , Masculino , Glicoproteínas de Membrana/genética , Camundongos , Microvilosidades/metabolismo , Dados de Sequência Molecular , Oócitos/metabolismo , Especificidade de Órgãos , Alinhamento de Sequência , Xenopus laevis
13.
J Biol Chem ; 274(46): 32889-96, 1999 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-10551853

RESUMO

An epithelial sodium channel (ENaC) is composed of three homologous subunits: alpha, beta, and gamma. To elucidate the function of the cytoplasmic, NH(2) terminus of rat ENaC (rENaC) subunits, a series of mutant cDNAs was constructed and the cRNAs for all three subunits were expressed in Xenopus oocytes. Amiloride-sensitive Na(+) currents (I(Na)) were measured by the two-electrode voltage clamp technique. Deletion of the cytoplasmic, NH(2) terminus of alpha (Delta2-109), beta (Delta2-49), or gamma-rENaC (Delta2-53) dramatically reduced I(Na). A series of progressive, NH(2)-terminal deletions of alpha-rENaC were constructed to identify motifs that regulate I(Na). Deletion of amino acids 2-46 had no effect on I(Na): however, deletion of amino acids 2-51, 2-55, 2-58, and 2-67 increased I(Na) by approximately 4-fold. By contrast, deletion of amino acids 2-79, 2-89, 2-100, and 2-109 eliminated I(Na). To evaluate the mechanism whereby Delta2-67-alpha-rENaC increased I(Na), single channels were evaluated by patch clamp. The single-channel conductance and open probability of alpha,beta,gamma-rENaC and Delta2-67-alpha,beta,gamma-rENaC were similar. However, the number of active channels in the membrane increased from 6 +/- 1 channels per patch with alpha,beta,gamma-rENaC to 11 +/- 1 channels per patch with Delta2-67-alpha,beta,gamma-rENaC. Laser scanning confocal microscopy confirmed that there were more Delta2-67-alpha,beta, gamma-rENaC channels in the plasma membrane than alpha,beta, gamma-rENaC channels. Deletion of amino acids 2-67 in alpha-rENaC reduced the endocytic retrieval of channels from the plasma membrane and increased the half-life of the channel in the membrane from 1.1 +/- 0.2 to 3.5 +/- 1.1 h. We conclude that the cytoplasmic, NH(2) terminus of alpha-, beta-, and gamma-rENaC is required for channel activity. The cytoplasmic, NH(2) terminus of alpha-rENaC contains two key motifs. One motif regulates the endocytic retrieval of the channel from the plasma membrane. The second motif is required for channel activity.


Assuntos
Endocitose/genética , Canais de Sódio/metabolismo , Amilorida/farmacologia , Animais , Brefeldina A/farmacologia , Membrana Celular/metabolismo , Canais Epiteliais de Sódio , Expressão Gênica , Proteínas de Fluorescência Verde , Proteínas Luminescentes , Microscopia Confocal , Oócitos , Técnicas de Patch-Clamp , RNA Complementar/genética , Ratos , Deleção de Sequência , Sódio/metabolismo , Canais de Sódio/genética , Xenopus
14.
J Clin Invest ; 104(7): 967-74, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10510337

RESUMO

The autosomal recessive form of type I pseudohypoaldosteronism (PHA-I) is an inherited salt-losing syndrome resulting from diminution-of-function mutations in the 3 subunits of the epithelial Na+ channel (ENaC). A PHA-I stop mutation (alpha(R508stop)) of the ENaC alpha subunit is predicted to lack the second transmembrane domain and the intracellular COOH-terminus, regions of the protein involved in pore function. Nonetheless, we observed a measurable Na+ current in Xenopus laevis oocytes that coexpress the beta and gamma subunits with the truncated alpha subunit. The mutant alpha was coassembled with beta and gamma subunits and was present at the cell surface at a lower density, consistent with the lower Na+ current seen in oocytes with the truncated alpha subunit. The single-channel Na+ conductance for the mutant channel was only slightly decreased, and the appearance of the macroscopic currents was delayed by 48 hours with respect to wild-type. Our data suggest novel roles for the alpha subunit in the assembly and targeting of an active channel to the cell surface, and suggest that channel pores consisting of only the beta and gamma subunits can provide significant residual activity. This activity may be sufficient to explain the absence of a severe pulmonary phenotype in patients with PHA-I.


Assuntos
Códon de Terminação , Hipoaldosteronismo/genética , Deleção de Sequência , Canais de Sódio/genética , Amilorida/farmacologia , Animais , Canais Epiteliais de Sódio , Feminino , Genes Recessivos , Humanos , Substâncias Macromoleculares , Mutagênese Sítio-Dirigida , Oócitos/fisiologia , Ratos , Proteínas Recombinantes/efeitos dos fármacos , Proteínas Recombinantes/metabolismo , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/fisiologia , Xenopus laevis
15.
Am J Physiol ; 277(4): L700-8, 1999 10.
Artigo em Inglês | MEDLINE | ID: mdl-10516210

RESUMO

Sodium 4-phenylbutyrate (PBA), a short-chain fatty acid, has been approved to treat patients with urea cycle enzyme deficiencies and is being evaluated in the management of sickle cell disease, thalassemia, cancer, and cystic fibrosis (CF). Because relatively little is known about the effects of PBA on the expression and function of the wild-type CF transmembrane conductance regulator (wt CFTR), the goal of this study was to examine the effects of PBA and related compounds on wt CFTR-mediated Cl(-) secretion. To this end, we studied Calu-3 cells, a human airway cell line that expresses endogenous wt CFTR and has a serous cell phenotype. We report that chronic treatment of Calu-3 cells with a high concentration (5 mM) of PBA, sodium butyrate, or sodium valproate but not of sodium acetate reduced basal and 8-(4-chlorophenylthio)-cAMP-stimulated Cl(-) secretion. Paradoxically, PBA enhanced CFTR protein expression 6- to 10-fold and increased the intensity of CFTR staining in the apical plasma membrane. PBA also increased protein expression of Na(+)-K(+)-ATPase. PBA reduced CFTR Cl(-) currents across the apical membrane but had no effect on Na(+)-K(+)-ATPase activity in the basolateral membrane. Thus a high concentration of PBA (5 mM) reduces Cl(-) secretion by inhibiting CFTR Cl(-) currents across the apical membrane. In contrast, lower therapeutic concentrations of PBA (0.05-2 mM) had no effect on cAMP-stimulated Cl(-) secretion across Calu-3 cells. We conclude that PBA concentrations in the therapeutic range are unlikely to have a negative effect on Cl(-) secretion. However, concentrations >5 mM might reduce transepithelial Cl(-) secretion by serous cells in submucosal glands in individuals expressing wt CFTR.


Assuntos
Cloretos/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Fenilbutiratos/farmacologia , Sistema Respiratório/metabolismo , Butiratos/farmacologia , Linhagem Celular , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Relação Dose-Resposta a Droga , Condutividade Elétrica , Células Epiteliais/metabolismo , Humanos , Fenilbutiratos/administração & dosagem , Sistema Respiratório/citologia , ATPase Trocadora de Sódio-Potássio/metabolismo , Ácido Valproico/farmacologia
16.
Am J Physiol ; 277(2): F271-6, 1999 08.
Artigo em Inglês | MEDLINE | ID: mdl-10444582

RESUMO

Sodium butyrate and its derivatives are useful therapeutic agents for the treatment of genetic diseases including urea cycle disorders, sickle cell disease, thalassemias, and possibly cystic fibrosis (CF). Butyrate partially restores cAMP-activated Cl(-) secretion in CF epithelial cells by stimulating DeltaF508 cystic fibrosis transmembrane conductance regulator (DeltaF508-CFTR) gene expression and increasing the amount of DeltaF508-CFTR in the plasma membrane. Because the effect of butyrate on Cl(-) secretion by renal epithelial cells has not been reported, we examined the effects of chronic butyrate treatment (15-18 h) on the function, expression, and localization of CFTR fused to the green fluorescent protein (GFP-CFTR) in stably transfected MDCK cells. We report that sodium butyrate reduced Cl(-) secretion across MDCK cells, yet increased apical membrane GFP-CFTR expression 25-fold and increased apical membrane Cl(-) currents 30-fold. Although butyrate also increased Na-K-ATPase protein expression twofold, the drug reduced the activity of the Na-K-ATPase by 55%. Our findings suggest that butyrate inhibits cAMP-stimulated Cl(-) secretion across MDCK cells in part by reducing the activity of the Na-K-ATPase.


Assuntos
Butiratos/farmacologia , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Rim/efeitos dos fármacos , Rim/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Cloretos/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cães , Condutividade Elétrica , Eletroquímica , Proteínas de Fluorescência Verde , Indicadores e Reagentes , Membranas Intracelulares/metabolismo , Rim/citologia , Proteínas Luminescentes/genética , Proteínas Recombinantes de Fusão/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo
17.
J Clin Invest ; 103(5): 667-73, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10074483

RESUMO

Liddle's syndrome is an inherited form of hypertension linked to mutations in the epithelial Na+ channel (ENaC). ENaC is composed of three subunits (alpha, beta, gamma), each containing a COOH-terminal PY motif (xPPxY). Mutations causing Liddle's syndrome alter or delete the PY motifs of beta- or gamma-ENaC. We recently demonstrated that the ubiquitin-protein ligase Nedd4 binds these PY motifs and that ENaC is regulated by ubiquitination. Here, we investigate, using the Xenopus oocyte system, whether Nedd4 affects ENaC function. Overexpression of wild-type Nedd4, together with ENaC, inhibited channel activity, whereas a catalytically inactive Nedd4 stimulated it, likely by acting as a competitive antagonist to endogenous Nedd4. These effects were dependant on the PY motifs, because no Nedd4-mediated changes in channel activity were observed in ENaC lacking them. The effect of Nedd4 on ENaC missing only one PY motif (of beta-ENaC), as originally described in patients with Liddle's syndrome, was intermediate. Changes were due entirely to alterations in ENaC numbers at the plasma membrane, as determined by surface binding and immunofluorescence. Our results demonstrate that Nedd4 is a negative regulator of ENaC and suggest that the loss of Nedd4 binding sites in ENaC observed in Liddle's syndrome may explain the increase in channel number at the cell surface, increased Na+ reabsorption by the distal nephron, and hence the hypertension.


Assuntos
Proteínas de Ligação ao Cálcio/metabolismo , Hipertensão/genética , Hipertensão/metabolismo , Ligases , Canais de Sódio/metabolismo , Ubiquitina-Proteína Ligases , Animais , Proteínas de Ligação ao Cálcio/genética , Complexos Endossomais de Distribuição Requeridos para Transporte , Células Epiteliais/metabolismo , Mutação , Ubiquitina-Proteína Ligases Nedd4 , Ratos , Canais de Sódio/genética , Síndrome , Xenopus , Proteínas de Xenopus
18.
Autoimmunity ; 28(3): 139-50, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9867126

RESUMO

MRL-Fas(lpr) mice spontaneously develop a chronic lupus-like renal disease, characterized by immune complex-mediated glomerulonephritis and abundant mononuclear cell infiltration in the interstitium. In the present study we have examined whether the macrophage chemoattractant osteopontin (Opn) could be important in the recruitment of macrophages in this murine model of autoimmune renal injury. We have examined the expression of Opn in the kidney of MRL-Fas(lpr) mice and have correlated Opn synthesis with the degree of macrophage infiltration. Immunofluorescence staining revealed prominent expression of Opn by proximal tubules in MRL-Fas(lpr) mice but not in MRL-++ control mice. Northern blot analysis demonstrated that steady-state transcript levels for Opn mRNA were also significantly increased in MRL-Fas(lpr) kidneys compared with control kidneys. Furthermore, in situ hybridization showed massive Opn mRNA transcripts in proximal tubules in MRL-Fas(lpr) mice but not in controls. The diffuse macrophage infiltration in the kidney of MRL-Fas(lpr) correlated with the enhanced Opn expression. Opn secretion in vitro by cultured renal tubular epithelial cells was upregulated by TNF-alpha and 1,25(OH)2-vitamin D3, whereas no regulation was observed in a control macrophage cell line. We conclude that the enhanced expression of the chemotactic molecule Opn by tubular cells is a prominent feature of murine lupus nephritis and might be promoted by the proinflammatory cytokine environment in MRL-Fas(lpr). The chronic upregulation of Opn could participate in the recruitment of monocytes in the kidney of MRL-Fas(lpr) mice, thereby contributing to the pathogenesis of autoimmune renal disease.


Assuntos
Rim/metabolismo , Nefrite Lúpica/metabolismo , Macrófagos/imunologia , Sialoglicoproteínas/biossíntese , Animais , Anticorpos Monoclonais/imunologia , Northern Blotting , Western Blotting , Modelos Animais de Doenças , Técnica Indireta de Fluorescência para Anticorpo , Hibridização In Situ , Túbulos Renais Proximais/metabolismo , Nefrite Lúpica/imunologia , Camundongos , Camundongos Endogâmicos CBA , Camundongos Endogâmicos MRL lpr , Osteopontina , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Sialoglicoproteínas/genética
19.
Mol Biol Cell ; 9(12): 3417-27, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9843578

RESUMO

The small G protein K-Ras2A is rapidly induced by aldosterone in A6 epithelia. In these Xenopus sodium reabsorbing cells, aldosterone rapidly activates preexisting epithelial Na+ channels (XENaC) via a transcriptionally mediated mechanism. In the Xenopus oocytes expression system, we tested whether the K-Ras2A pathway impacts on XENaC activity by expressing XENaC alone or together with XK-Ras2A rendered constitutively active (XK-Ras2AG12V). As a second control, XENaC-expressing oocytes were treated with progesterone, a sex steroid that induces maturation of the oocytes similarly to activated Ras. Progesterone or XK-Ras2AG12V led to oocyte maturation characterized by a decrease in surface area and endogenous Na+ pump function. In both conditions, the surface expression of exogenous XENaC's was also decreased; however, in comparison with progesterone-treated oocytes, XK-ras2AG12V-coinjected oocytes expressed a fivefold higher XENaC-mediated macroscopic Na+ current that was as high as that of control oocytes. Thus, the Na+ current per surface-expressed XENaC was increased by XK-Ras2AG12V. The chemical driving force for Na+ influx was not changed, suggesting that XK-Ras2AG12V increased the mean activity of XENaCs at the oocyte surface. These observations raise the possibility that XK-Ras2A, which is the first regulatory protein known to be transcriptionally induced by aldosterone, could play a role in the control of XENaC function in aldosterone target cells.


Assuntos
Canais de Sódio/metabolismo , Proteínas ras/metabolismo , Aldosterona/farmacologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Técnicas In Vitro , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Progesterona/farmacologia , Canais de Sódio/efeitos dos fármacos , Canais de Sódio/genética , Xenopus , Proteínas ras/genética
20.
FEBS Lett ; 439(1-2): 157-62, 1998 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-9849898

RESUMO

The protein mediating system L amino acid transport, AmAT-L, is a disulfide-linked heterodimer of a permease-related light chain (AmAT-L-lc) and the type II glycoprotein 4F2hc/ CD98. The Schistosoma mansoni protein SPRM1 also heterodimerizes with h4F2hc, inducing amino acid transport with different specificity. In this study, we show that the disulfide bond is formed by heavy chain C109 with a Cys residue located in the second putative extracellular loop of the multi-transmembrane domain light chain (C164 and C137 for XAmAT-L-lc and SPRM1, respectively). The non-covalent interaction of Cys-mutant subunits is not sufficient to allow coimmunoprecipitation, but cell surface expression of the light chains is maintained to a large extent. The non-covalently linked transporters display the same transport characteristics as disulfide bound heterodimers, but the maximal transport rates are reduced by 30-80%.


Assuntos
Proteínas de Transporte/metabolismo , Cisteína/metabolismo , Dissulfetos/metabolismo , Sistemas de Transporte de Aminoácidos , Animais , Proteínas de Transporte/química , Proteínas de Transporte/genética , Dimerização , Mutagênese Sítio-Dirigida , Oócitos/metabolismo , Testes de Precipitina , Conformação Proteica , Relação Estrutura-Atividade , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...