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.
Physiol Rev ; 95(1): 297-340, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25540145

RESUMO

Transcription and translation require a high concentration of potassium across the entire tree of life. The conservation of a high intracellular potassium was an absolute requirement for the evolution of life on Earth. This was achieved by the interplay of P- and V-ATPases that can set up electrochemical gradients across the cell membrane, an energetically costly process requiring the synthesis of ATP by F-ATPases. In animals, the control of an extracellular compartment was achieved by the emergence of multicellular organisms able to produce tight epithelial barriers creating a stable extracellular milieu. Finally, the adaptation to a terrestrian environment was achieved by the evolution of distinct regulatory pathways allowing salt and water conservation. In this review we emphasize the critical and dual role of Na(+)-K(+)-ATPase in the control of the ionic composition of the extracellular fluid and the renin-angiotensin-aldosterone system (RAAS) in salt and water conservation in vertebrates. The action of aldosterone on transepithelial sodium transport by activation of the epithelial sodium channel (ENaC) at the apical membrane and that of Na(+)-K(+)-ATPase at the basolateral membrane may have evolved in lungfish before the emergence of tetrapods. Finally, we discuss the implication of RAAS in the origin of the present pandemia of hypertension and its associated cardiovascular diseases.


Assuntos
Aldosterona/metabolismo , Evolução Biológica , Canais Epiteliais de Sódio/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sódio/metabolismo , Animais , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Genoma Humano , Humanos , Néfrons/fisiologia , Transdução de Sinais/fisiologia , ATPase Trocadora de Sódio-Potássio/química , ATPase Trocadora de Sódio-Potássio/genética
3.
Pflugers Arch ; 462(6): 871-83, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21938401

RESUMO

Inositol Inpp5k (or Pps, SKIP) is a member of the inositol polyphosphate 5-phosphatases family with a poorly characterized function in vivo. In this study, we explored the function of this inositol 5-phosphatase in mice and cells overexpressing the 42-kDa mouse Inpp5k protein. Inpp5k transgenic mice present defects in water metabolism characterized by a reduced plasma osmolality at baseline, a delayed urinary water excretion following a water load, and an increased acute response to vasopressin. These defects are associated with the expression of the Inpp5k transgene in renal collecting ducts and with alterations in the arginine vasopressin/aquaporin-2 signalling pathway in this tubular segment. Analysis in a mouse collecting duct mCCD cell line revealed that Inpp5k overexpression leads to increased expression of the arginine vasopressin receptor type 2 and increased cAMP response to arginine vasopressin, providing a basis for increased aquaporin-2 expression and plasma membrane localization with increased osmotically induced water transport. Altogether, our results indicate that Inpp5k 5-phosphatase is important for the control of the arginine vasopressin/aquaporin-2 signalling pathway and water transport in kidney collecting ducts.


Assuntos
Aquaporina 2/metabolismo , Túbulos Renais Coletores/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Vasopressinas/metabolismo , Equilíbrio Hidroeletrolítico/fisiologia , Animais , Células Cultivadas , Feminino , Humanos , Túbulos Renais Coletores/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Monoéster Fosfórico Hidrolases/genética , Transdução de Sinais/fisiologia , Água/metabolismo
4.
Physiol Genomics ; 43(13): 844-54, 2011 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-21558422

RESUMO

Despite large changes in salt intake, the mammalian kidney is able to maintain the extracellular sodium concentration and osmolarity within very narrow margins, thereby controlling blood volume and blood pressure. In the aldosterone-sensitive distal nephron (ASDN), aldosterone tightly controls the activities of epithelial sodium channel (ENaC) and Na,K-ATPase, the two limiting factors in establishing transepithelial sodium transport. It has been proposed that the ENaC/degenerin gene family is restricted to Metazoans, whereas the α- and ß-subunits of Na,K-ATPase have homologous genes in prokaryotes. This raises the question of the emergence of osmolarity control. By exploring recent genomic data of diverse organisms, we found that: 1) ENaC/degenerin exists in all of the Metazoans screened, including nonbilaterians and, by extension, was already present in ancestors of Metazoa; 2) ENaC/degenerin is also present in Naegleria gruberi, an eukaryotic microbe, consistent with either a vertical inheritance from the last common ancestor of Eukaryotes or a lateral transfer between Naegleria and Metazoan ancestors; and 3) The Na,K-ATPase ß-subunit is restricted to Holozoa, the taxon that includes animals and their closest single-cell relatives. Since the ß-subunit of Na,K-ATPase plays a key role in targeting the α-subunit to the plasma membrane and has an additional function in the formation of cell junctions, we propose that the emergence of Na,K-ATPase, together with ENaC/degenerin, is linked to the development of multicellularity in the Metazoan kingdom. The establishment of multicellularity and the associated extracellular compartment ("internal milieu") precedes the emergence of other key elements of the aldosterone signaling pathway.


Assuntos
Aldosterona/metabolismo , Canais Epiteliais de Sódio/genética , Evolução Molecular , ATPase Trocadora de Sódio-Potássio/genética , Sódio/metabolismo , Canais Iônicos Sensíveis a Ácido , Animais , Canais de Sódio Degenerina , Humanos , Transporte de Íons/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/genética , Fosfoproteínas/genética , Filogenia
5.
J Am Soc Nephrol ; 22(2): 253-61, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21051735

RESUMO

Lithium-induced nephrogenic diabetes insipidus (NDI) is accompanied by polyuria, downregulation of aquaporin 2 (AQP2), and cellular remodeling of the collecting duct (CD). The amiloride-sensitive epithelial sodium channel (ENaC) is a likely candidate for lithium entry. Here, we subjected transgenic mice lacking αENaC specifically in the CD (knockout [KO] mice) and littermate controls to chronic lithium treatment. In contrast to control mice, KO mice did not markedly increase their water intake. Furthermore, KO mice did not demonstrate the polyuria and reduction in urine osmolality induced by lithium treatment in the control mice. Lithium treatment reduced AQP2 protein levels in the cortex/outer medulla and inner medulla (IM) of control mice but only partially reduced AQP2 levels in the IM of KO mice. Furthermore, lithium induced expression of H(+)-ATPase in the IM of control mice but not KO mice. In conclusion, the absence of functional ENaC in the CD protects mice from lithium-induced NDI. These data support the hypothesis that ENaC-mediated lithium entry into the CD principal cells contributes to the pathogenesis of lithium-induced NDI.


Assuntos
Diabetes Insípido Nefrogênico/induzido quimicamente , Canais Epiteliais de Sódio/fisiologia , Cloreto de Lítio/toxicidade , Absorção , Animais , Aquaporina 2/análise , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/patologia , Cloreto de Lítio/farmacocinética , Camundongos , Camundongos Knockout , ATPases Translocadoras de Prótons/análise
6.
J Biol Chem ; 285(35): 26945-26955, 2010 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-20566636

RESUMO

Studies in cystic fibrosis patients and mice overexpressing the epithelial Na(+) channel beta-subunit (betaENaC-Tg) suggest that raised airway Na(+) transport and airway surface liquid (ASL) depletion are central to the pathogenesis of cystic fibrosis lung disease. However, patients or mice with Liddle gain-of-function betaENaC mutations exhibit hypertension but no lung disease. To investigate this apparent paradox, we compared the airway phenotype (nasal versus tracheal) of Liddle with CFTR-null, betaENaC-Tg, and double mutant mice. In mouse nasal epithelium, the region that functionally mimics human airways, high levels of CFTR expression inhibited Liddle epithelial Nat channel (ENaC) hyperfunction. Conversely, in mouse trachea, low levels of CFTR failed to suppress Liddle ENaC hyperfunction. Indeed, Na(+) transport measured in Ussing chambers ("flooded" conditions) was raised in both Liddle and betaENaC-Tg mice. Because enhanced Na(+) transport did not correlate with lung disease in these mutant mice, measurements in tracheal cultures under physiologic "thin film" conditions and in vivo were performed. Regulation of ASL volume and ENaC-mediated Na(+) absorption were intact in Liddle but defective in betaENaC-Tg mice. We conclude that the capacity to regulate Na(+) transport and ASL volume, not absolute Na(+) transport rates in Ussing chambers, is the key physiologic function protecting airways from dehydration-induced lung disease.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Canais Epiteliais de Sódio/biossíntese , Regulação da Expressão Gênica , Síndrome de Liddle/metabolismo , Mutação , Sódio/metabolismo , Animais , Fibrose Cística/metabolismo , Fibrose Cística/patologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Canais Epiteliais de Sódio/genética , Humanos , Transporte de Íons/genética , Síndrome de Liddle/patologia , Camundongos , Camundongos Knockout , Mucosa Nasal/metabolismo , Mucosa Nasal/patologia , Técnicas de Cultura de Órgãos , Traqueia/metabolismo , Traqueia/patologia
7.
EMBO Mol Med ; 2(1): 26-37, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20043279

RESUMO

Sodium transport via epithelial sodium channels (ENaC) expressed in alveolar epithelial cells (AEC) provides the driving force for removal of fluid from the alveolar space. The membrane-bound channel-activating protease 1 (CAP1/Prss8) activates ENaC in vitro in various expression systems. To study the role of CAP1/Prss8 in alveolar sodium transport and lung fluid balance in vivo, we generated mice lacking CAP1/Prss8 in the alveolar epithelium using conditional Cre-loxP-mediated recombination. Deficiency of CAP1/Prss8 in AEC induced in vitro a 40% decrease in ENaC-mediated sodium currents. Sodium-driven alveolar fluid clearance (AFC) was reduced in CAP1/Prss8-deficient mice, due to a 48% decrease in amiloride-sensitive clearance, and was less sensitive to beta(2)-agonist treatment. Intra-alveolar treatment with neutrophil elastase, a soluble serine protease activating ENaC at the cell surface, fully restored basal AFC and the stimulation by beta(2)-agonists. Finally, acute volume-overload increased alveolar lining fluid volume in CAP1/Prss8-deficient mice. This study reveals that CAP1 plays a crucial role in the regulation of ENaC-mediated alveolar sodium and water transport and in mouse lung fluid balance.


Assuntos
Canais Epiteliais de Sódio/metabolismo , Água Extravascular Pulmonar/metabolismo , Pulmão/metabolismo , Alvéolos Pulmonares/metabolismo , Serina Endopeptidases/metabolismo , Equilíbrio Hidroeletrolítico , Agonistas Adrenérgicos beta/farmacologia , Animais , Células Cultivadas , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/genética , Deleção de Genes , Expressão Gênica , Camundongos , Alvéolos Pulmonares/citologia , Edema Pulmonar/metabolismo , Mucosa Respiratória/metabolismo , Serina Endopeptidases/genética , Equilíbrio Hidroeletrolítico/efeitos dos fármacos
8.
Am J Physiol Renal Physiol ; 296(2): F446-57, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19052103

RESUMO

Polycystic kidney diseases result from disruption of the genetically defined program that controls the size and geometry of renal tubules. Cysts which frequently arise from the collecting duct (CD) result from cell proliferation and fluid secretion. From mCCD(cl1) cells, a differentiated mouse CD cell line, we isolated a clonal subpopulation (mCCD-N21) that retains morphogenetic capacity. When grown in three-dimensional gels, mCCD-N21 cells formed highly organized tubular structures consisting of a palisade of polarized epithelial cells surrounding a cylindrical lumen. Subsequent addition of cAMP-elevating agents (forskolin or cholera toxin) or of membrane-permeable cAMP analogs (CPT-cAMP) resulted in rapid and progressive dilatation of existing tubules, leading to the formation of cystlike structures. When grown on filters, mCCD-N21 cells exhibited a high transepithelial resistance as well as aldosterone- and/or vasopressin-induced amiloride-sensitive and -insensitive current. The latter was in part inhibited by Na(+)-K(+)-2Cl(-) cotransporter (bumetanide) and chloride channel (NPPB) inhibitors. Real-time PCR analysis confirmed the expression of NKCC1, the ubiquitous Na(+)-K(+)-2Cl(-) cotransporter and cystic fibrosis transmembrane regulator (CFTR) in mCCD-N21 cells. Tubule enlargement and cyst formation were prevented by inhibitors of Na(+)-K(+)-2Cl(-) cotransporters (bumetanide or ethacrynic acid) or CFTR (NPPB or CFTR inhibitor-172). These results further support the notion that cAMP signaling plays a key role in renal cyst formation, at least in part by promoting chloride-driven fluid secretion. This new in vitro model of tubule-to-cyst conversion affords a unique opportunity for investigating the molecular mechanisms that govern the architecture of epithelial tubes, as well as for dissecting the pathophysiological processes underlying cystic kidney diseases.


Assuntos
Técnicas de Cultura de Células , Cloretos/metabolismo , AMP Cíclico/metabolismo , Túbulos Renais Coletores/citologia , Animais , Linhagem Celular , Doenças Renais Císticas/patologia , Camundongos
9.
J Am Soc Nephrol ; 18(6): 1672-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17475820

RESUMO

The antidiuretic effect of vasopressin is mediated by V2 receptors (V2R) that are located in kidney connecting tubules and collecting ducts. This study provides evidence that V2R signaling is negatively regulated by regulator of G protein signaling 2 (RGS2), a member of the family of RGS proteins. This study demonstrates that (1) RGS2 expression in the kidney is restricted to the vasopressin-sensitive part of the nephron (thick ascending limb, connecting tubule, and collecting duct); (2) expression of RGS2 is rapidly upregulated by vasopressin; (3) the vasopressin-dependent accumulation of cAMP, the principal messenger of V2R signaling, is significantly higher in collecting ducts that are microdissected from the RGS2(-/-) mice compared with their wild-type littermates; and (4) analysis of urine output of mice that were exposed to water restriction followed by acute water loading revealed that RGS2(-/-) mice exhibit an increased renal responsiveness to vasopressin. It is proposed that RGS2 is involved in negative feedback regulation of V2R signaling.


Assuntos
Antidiuréticos/farmacologia , Túbulos Renais Coletores/metabolismo , Proteínas RGS/genética , Receptores de Vasopressinas/metabolismo , Transdução de Sinais/fisiologia , Vasopressinas/farmacologia , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Ingestão de Líquidos/fisiologia , Endotelina-1/farmacologia , Retroalimentação Fisiológica/efeitos dos fármacos , Retroalimentação Fisiológica/fisiologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Néfrons/efeitos dos fármacos , Néfrons/metabolismo , Fenilefrina/farmacologia , Proteínas RGS/metabolismo , RNA Mensageiro/metabolismo , Receptores de Vasopressinas/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Sódio/sangue , Sódio/urina , Simpatomiméticos/farmacologia , Privação de Água/fisiologia
10.
J Physiol ; 582(Pt 2): 777-88, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17430990

RESUMO

Transepithelial sodium transport via alveolar epithelial Na(+) channels and Na(+),K(+)-ATPase constitutes the driving force for removal of alveolar oedema fluid. Decreased activity of the amiloride-sensitive epithelial Na(+) channel (ENaC) in the apical membrane of alveolar epithelial cells impairs sodium-driven alveolar fluid clearance (AFC) and predisposes to pulmonary oedema. We hypothesized that hyperactivity of ENaC in the distal lung could improve AFC and facilitate the resolution of pulmonary oedema. AFC and lung fluid balance were studied at baseline and under conditions of hydrostatic pulmonary oedema in the beta-Liddle (L) mouse strain harbouring a gain-of-function mutation (R(566)(stop)) within the Scnn1b gene. As compared with wild-type (+/+), baseline AFC was increased by 2- and 3-fold in heterozygous (+/L) and homozygous mutated (L/L) mice, respectively, mainly due to increased amiloride-sensitive AFC. The beta(2)-agonist terbutaline stimulated AFC in +/+ and +/L mice, but not in L/L mice. Acute volume overload induced by saline infusion (40% of body weight over 2 h) significantly increased extravascular (i.e. interstitial and alveolar) lung water as assessed by the bloodless wet-to-dry lung weight ratio in +/+ and L/L mice, as compared with baseline. However, the increase was significantly larger in +/+ than in L/L groups (P=0.01). Volume overload also increased the volume of the alveolar epithelial lining fluid in +/+ mice, indicating the presence of alveolar oedema, but not in L/L mice. Cardiac function as evaluated by echocardiography was comparable in both groups. These data show that constitutive ENaC activation improved sodium-driven AFC in the mouse lung, and attenuated the severity of hydrostatic pulmonary oedema.


Assuntos
Líquidos Corporais/metabolismo , Canais Epiteliais de Sódio/genética , Mutação , Alvéolos Pulmonares/metabolismo , Edema Pulmonar/fisiopatologia , Agonistas Adrenérgicos beta/farmacologia , Amilorida/farmacologia , Animais , Brônquios/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/antagonistas & inibidores , Diuréticos/farmacologia , Canais Epiteliais de Sódio/metabolismo , Glicina/análogos & derivados , Glicina/farmacologia , Coração/fisiopatologia , Pressão Hidrostática , Pulmão/patologia , Camundongos , Camundongos Mutantes , Camundongos Transgênicos , Isoformas de Proteínas/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Edema Pulmonar/genética , Edema Pulmonar/patologia , Índice de Gravidade de Doença , Terbutalina/farmacologia
11.
J Biol Chem ; 282(1): 58-64, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17090546

RESUMO

The amiloride-sensitive epithelial sodium channel (ENaC) constitutes a limiting step in sodium reabsorption across distal airway epithelium and controlling mucociliary clearance. ENaC is activated by serine proteases secreted in the extracellular milieu. In cystic fibrosis lungs, high concentrations of secreted neutrophil elastase (NE) are observed. hNE could activate ENaC and contribute to further decreased mucociliary clearance. The aims of this study were (i) to test the ability of an engineered human neutrophil elastase inhibitor (EPI-hNE4) to specifically inhibit the elastase activation of ENaC-mediated amiloride-sensitive currents (I(Na)) and (ii) to examine the effect of elastase on cell surface expression of ENaC and its cleavage pattern (exogenous proteolysis). Oocytes were exposed to hNE (10-100 microg/ml) and/or trypsin (10 microg/ml) for 2-5 min in the presence or absence of EPI-hNE4 (0.7 microm). hNE activated I(Na) 3.6-fold (p < 0.001) relative to non-treated hENaC-injected oocytes. EPI-hNE4 fully inhibited hNE-activated I(Na) but had no effect on trypsin- or prostasin-activated I(Na). The co-activation of I(Na) by hNE and trypsin was not additive. Biotinylation experiments revealed that cell surface gamma ENaC (but not alpha or beta ENaC) exposed to hNE for 2 min was cleaved (as a 67-kDa fragment) and correlated with increased I(Na). The elastase-induced exogenous proteolysis pattern is distinct from the endogenous proteolysis pattern induced upon preferential assembly, suggesting a causal relationship between gamma ENaC cleavage and ENaC activation, taking place at the plasma membrane.


Assuntos
Canais Epiteliais de Sódio/fisiologia , Elastase de Leucócito/antagonistas & inibidores , Oócitos/metabolismo , Peptídeos/farmacologia , Animais , Membrana Celular/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/química , Canais Epiteliais de Sódio/metabolismo , Peptídeos/química , Engenharia de Proteínas , RNA Complementar/metabolismo , Serina Endopeptidases/química , Propriedades de Superfície , Tripsina/química , Xenopus laevis
12.
J Biol Chem ; 280(46): 38264-70, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16172119

RESUMO

Activation of the mitogen-activated protein (MAP) kinase cascade by progesterone in Xenopus oocytes leads to a marked down-regulation of activity of the amiloride-sensitive epithelial sodium channel (ENaC). Here we have studied the signaling pathways involved in progesterone effect on ENaC activity. We demonstrate that: (i) the truncation of the C termini of the alphabetagammaENaC subunits results in the loss of the progesterone effect on ENaC; (ii) the effect of progesterone was also suppressed by mutating conserved tyrosine residues in the Pro-X-X-Tyr (PY) motif of the C termini of the beta and gamma ENaC subunits (beta(Y618A) and gamma(Y628A)); (iii) the down-regulation of ENaC activity by progesterone was also suppressed by co-expression ENaC subunits with a catalytically inactive mutant of Nedd4-2, a ubiquitin ligase that has been previously demonstrated to decrease ENaC cell-surface expression via a ubiquitin-dependent internalization/degradation mechanism; (iv) the effect of progesterone was significantly reduced by suppression of consensus sites (beta(T613A) and gamma(T623A)) for ENaC phosphorylation by the extracellular-regulated kinase (ERK), a MAP kinase previously shown to facilitate the binding of Nedd4 ubiquitin ligases to ENaC; (v) the quantification of cell-surface-expressed ENaC subunits revealed that progesterone decreases ENaC open probability (whole cell P(o), wcP(o)) and not its cell-surface expression. Collectively, these results demonstrate that the binding of active Nedd4-2 to ENaC is a crucial step in the mechanism of ENaC inhibition by progesterone. Upon activation of ERK, the effect of Nedd4-2 on ENaC open probability can become more important than its effect on ENaC cell-surface expression.


Assuntos
Amilorida/farmacologia , Regulação para Baixo , Progesterona/fisiologia , Canais de Sódio/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/fisiologia , Ubiquitina-Proteína Ligases/fisiologia , Aldosterona/farmacologia , Motivos de Aminoácidos , Animais , Sítios de Ligação , Biotinilação , Catálise , Membrana Celular/metabolismo , Complexos Endossomais de Distribuição Requeridos para Transporte , Canais Epiteliais de Sódio , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases , Mutação , Ubiquitina-Proteína Ligases Nedd4 , Oócitos/metabolismo , Oxigênio/metabolismo , Técnicas de Patch-Clamp , Fosforilação , Progesterona/química , Progesterona/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , RNA Complementar/metabolismo , Ratos , Transdução de Sinais , Fatores de Tempo , Tirosina/química , Ubiquitina/química , Xenopus , Proteínas de Xenopus
13.
Hum Genet ; 117(6): 528-35, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16021470

RESUMO

Pathogenic mutations in TMPRSS3, which encodes a transmembrane serine protease, cause non-syndromic deafness DFNB8/10. Missense mutations map in the low density-lipoprotein receptor A (LDLRA), scavenger-receptor cysteine-rich (SRCR), and protease domains of the protein, indicating that all domains are important for its function. TMPRSS3 undergoes proteolytic cleavage and activates the ENaC sodium channel in a Xenopus oocyte model system. To assess the importance of this gene in non-syndromic childhood or congenital deafness in Turkey, we screened for mutations affected members of 25 unrelated Turkish families. The three families with the highest LOD score for linkage to chromosome 21q22.3 were shown to harbor P404L, R216L, or Q398X mutations, suggesting that mutations in TMPRSS3 are a considerable contributor to non-syndromic deafness in the Turkish population. The mutant TMPRSS3 harboring the novel R216L missense mutation within the predicted cleavage site of the protein fails to undergo proteolytic cleavage and is unable to activate ENaC, thus providing evidence that pre-cleavage of TMPRSS3 is mandatory for normal function.


Assuntos
Surdez/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Sequência de Aminoácidos , Cromossomos Humanos Par 21 , Feminino , Ligação Genética , Haplótipos , Humanos , Escore Lod , Masculino , Proteínas de Membrana/metabolismo , Dados de Sequência Molecular , Proteínas de Neoplasias/metabolismo , Linhagem , Serina Endopeptidases/metabolismo
14.
J Physiol ; 562(Pt 1): 271-84, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15513933

RESUMO

Liddle's syndrome is a genetic form of hypertension linked to Na(+) retention caused by activating mutations in the COOH terminus of the beta or gamma subunit of the epithelial sodium channel (ENaC). In this study, we used the short-circuit current (I(sc)) method to investigate the effects of deamino-8-d-arginine vasopressin (dDAVP) on Na(+) and Cl(-) fluxes in primary cultures of cortical collecting ducts (CCDs) microdissected from the kidneys of mice with Liddle's syndrome carrying a stop codon mutation, corresponding to the beta-ENaC R(566) stop mutation (L) found in the original pedigree. Compared to wild-type (+/+) CCD cells, untreated L/+ and L/L CCD cells exhibited 2.7- and 4.2-fold increases, respectively, in amiloride-sensitive (Ams) I(sc), reflecting ENaC-dependent Na(+) absorption. Short-term incubation with dDAVP caused a rapid and significant increase (approximately 2-fold) in Ams I(sc) in +/+, but not in L/+ or L/L CCD cells. In sharp contrast, dDAVP induced a greater increase in 5-nitro-2-(3-phenylpropamino)benzoate (NPPB)-inhibited apical Cl(-) currents in amiloride-treated L/L and L/+ cells than in their +/+ counterparts. I(sc) recordings performed under apical ion substituted conditions revealed that the dDAVP-stimulated apical secretion of Cl(-), which was absent in cultured CCDs lacking CFTR, was 1.8-fold greater in L/+ and 3.7-fold greater in L/L CCD cells than in their +/+ CCD counterparts. After the basal membrane had been permeabilized with nystatin and a basal-to-apical Cl(-) gradient had been imposed, dDAVP also stimulated larger Cl(-) currents across L/L and L/+ CCD layers than +/+ CCD layers. These findings demonstrate that vasopressin stimulates greater apical CFTR Cl(-) conductance in the renal CCD cells of mice with Liddle's syndrome than in wild-type mice. This effect could contribute to the enhanced NaCl reabsorption observed in the distal nephron of patients with Liddle's syndrome.


Assuntos
Agonistas dos Canais de Cloreto , Regulador de Condutância Transmembrana em Fibrose Cística/fisiologia , Hipertensão/genética , Hipertensão/metabolismo , Túbulos Renais Coletores/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Vasopressinas/farmacologia , Animais , Células Cultivadas , Canais de Cloreto/genética , Cloretos/metabolismo , Códon/genética , Desamino Arginina Vasopressina/metabolismo , Desamino Arginina Vasopressina/farmacologia , Eletrofisiologia , Canais Epiteliais de Sódio , Túbulos Renais Coletores/efeitos dos fármacos , Camundongos , Camundongos Knockout , Néfrons/metabolismo , Nistatina/farmacologia , Técnicas de Cultura de Órgãos , Fenótipo , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Canais de Sódio/genética , Síndrome
15.
EMBO J ; 23(22): 4517-25, 2004 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-15510217

RESUMO

The interleukin-6 cytokines, acting via gp130 receptor pathways, play a pivotal role in the reduction of cardiac injury induced by mechanical stress or ischemia and in promoting subsequent adaptive remodeling of the heart. We have now identified the small proline-rich repeat proteins (SPRR) 1A and 2A as downstream targets of gp130 signaling that are strongly induced in cardiomyocytes responding to biomechanical/ischemic stress. Upregulation of SPRR1A and 2A was markedly reduced in the gp130 cardiomyocyte-restricted knockout mice. In cardiomyocytes, MEK1/2 inhibitors prevented SPRR1A upregulation by gp130 cytokines. Furthermore, binding of NF-IL6 (C/EBPbeta) and c-Jun to the SPRR1A promoter was observed after CT-1 stimulation. Histological analysis revealed that SPRR1A induction after mechanical stress of pressure overload was restricted to myocytes surrounding piecemeal necrotic lesions. A similar expression pattern was found in postinfarcted rat hearts. Both in vitro and in vivo ectopic overexpression of SPRR1A protected cardiomyocytes against ischemic injury. Thus, this study identifies SPRR1A as a novel stress-inducible downstream mediator of gp130 cytokines in cardiomyocytes and documents its cardioprotective effect against ischemic stress.


Assuntos
Antígenos CD/metabolismo , Cardiotônicos/metabolismo , Glicoproteínas de Membrana/metabolismo , Traumatismo por Reperfusão Miocárdica , Proteínas/metabolismo , Adenoviridae/genética , Animais , Animais Recém-Nascidos , Western Blotting , Sobrevivência Celular , Proteínas Ricas em Prolina do Estrato Córneo , Cruzamentos Genéticos , Receptor gp130 de Citocina , Corantes Fluorescentes , Regulação da Expressão Gênica , Hidrazinas , Interleucina-6/genética , Interleucina-6/metabolismo , Proteínas de Membrana , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Biológicos , Miocárdio/citologia , Miocárdio/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas , Transdução de Sinais
16.
J Biol Chem ; 279(49): 51002-12, 2004 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-15456767

RESUMO

The collecting duct of normal kidney exhibits significant activity of the MEK1/2-ERK1/2 pathway as shown in vivo by immunostaining of phosphorylated active ERK1/2 (pERK1/2). The MEK1/2-ERK1/2 pathway controls many different ion transports both in proximal and distal nephron, raising the question of whether this pathway is involved in the basal and/or hormone-dependent transepithelial sodium reabsorption in the principal cell of the cortical collecting duct (CCD), a process mediated by the apical epithelial sodium channel and the basolateral sodium pump (Na,K-ATPase). To answer this question we used ex vivo microdissected CCDs from normal mouse kidney or in vitro cultured mpkCCDcl4 principal cells. Significant basal levels of pERK1/2 were observed ex vivo and in vitro. Aldosterone and vasopressin, known to up-regulate sodium reabsorption in CCDs, did not change ERK1/2 activity either ex vivo or in vitro. Basal and aldosterone- or vasopressin-stimulated sodium transport was down-regulated by the MEK1/2 inhibitor PD98059, in parallel with a decrease in pERK1/2 in vitro. The activity of Na,K-ATPase but not that of epithelial sodium channel was inhibited by MEK1/2 inhibitors in both unstimulated and aldosterone- or vasopressin-stimulated CCDs in vitro. Cell surface biotinylation showed that intrinsic activity rather than cell surface expression of Na,K-ATPase was controlled by pERK1/2. PD98059 also significantly inhibited the activity of Na,K-ATPase ex vivo. Our data demonstrate that the ERK1/2 pathway controls Na,K-ATPase activity and transepithelial sodium transport in the principal cell and indicate that basal constitutive activity of the ERK1/2 pathway is a critical component of this control.


Assuntos
Túbulos Renais Coletores/metabolismo , Rim/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/fisiologia , Proteína Quinase 3 Ativada por Mitógeno/fisiologia , ATPase Trocadora de Sódio-Potássio/química , Sódio/metabolismo , Aldosterona/metabolismo , Animais , Transporte Biológico , Biotinilação , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Ligantes , Masculino , Camundongos , Néfrons/metabolismo , Fosforilação , ATPase Trocadora de Sódio-Potássio/metabolismo , Fatores de Tempo , Regulação para Cima , Vasopressinas/metabolismo
17.
Biochemistry ; 42(34): 10333-41, 2003 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-12939163

RESUMO

The calcitonin receptor-like receptor (CRLR) and receptor activity modifying protein-3 (RAMP3) can assemble into a CRLR/RAMP3 heterodimeric receptor that exhibits the characteristics of a high affinity adrenomedullin receptor. RAMP3 participates in adrenomedullin (AM) binding via its extracellular N-terminus characterized by the presence of six highly conserved cysteine residues and four N-glycosylation consensus sites. Here, we assessed the usage of these conserved residues in cotranslational modifications of RAMP3 and addressed their role in functional expression of the CRLR/RAMP3 receptor. Using a Xenopus oocyte expression system, we show that (i) RAMP3 is assembled with CRLR as a multiple N-glycosylated species in which two, three, or four consensus sites are used; (ii) elimination of all N-glycans in RAMP3 results in a significant inhibition of receptor [(125)I]AM binding and an increase in the EC(50) value for AM; (iii) several lines of indirect evidence indicate that each of the six cysteines is involved in disulfide bond formation; (iv) when all cysteines are mutated to serines, RAMP3 is N-glycosylated at all four consensus sites, suggesting that disulfide bond formation inhibits N-gylcosylation; and (v) elimination of all cysteines abolishes adrenomedullin binding and leads to a complete loss of receptor function. Our data demonstrate that cotranslational modifications of RAMP3 play a critical role in the function of the CRLR/RAMP3 adrenomedullin receptor.


Assuntos
Cisteína/genética , Cisteína/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Receptores da Calcitonina/metabolismo , Receptores de Peptídeos/metabolismo , Adrenomedulina , Substituição de Aminoácidos , Animais , Sítios de Ligação , Peptídeo Relacionado com Gene de Calcitonina/metabolismo , Proteína Semelhante a Receptor de Calcitonina , Sequência Consenso , Dissulfetos/metabolismo , Glicosilação , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/genética , Oócitos/metabolismo , Peptídeos/metabolismo , Polissacarídeos/química , Ligação Proteica , Estrutura Terciária de Proteína , Ensaio Radioligante , Proteínas Modificadoras da Atividade de Receptores , Receptores de Adrenomedulina , Receptores da Calcitonina/química , Receptores de Peptídeos/química , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Xenopus
18.
Am J Physiol Renal Physiol ; 285(2): F310-8, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12684224

RESUMO

Currents through epithelial Na channels (ENaCs) were measured in the cortical collecting tubule (CCT) of mice expressing truncated beta-subunits of ENaC, reproducing one of the mutations found in human patients with Liddle's syndrome. Tubules were isolated from mice homozygous for the Liddle mutation (L/L) and from wild-type (WT) littermates. Amiloride-sensitive currents (INa) from single cells were recorded under whole cell clamp conditions. CCTs from mice kept under control conditions and fed a diet with normal levels of Na had very small INas (WT: 18 +/- 13 pA; L/L: 22 +/- 8 pA at Vm = -100 mV) that were not different in WT and L/L animals. However, the L/L mice had much larger currents when the animals were fed a low-Na diet (WT: 256 +/- 127 pA; L/L: 1,820 +/- 330 pA) or infused with aldosterone (WT: 285 +/- 63 pA; L/L: 1,600 +/- 280 pA). Currents from L/L mice were also larger when animals were pretreated with a high-K diet but not when the CCTs were stimulated in vitro with 8-CTP-cAMP. Noise analysis of amiloride-induced fluctuations in INa showed that single-channel currents at Vm = 0 mV were slightly smaller in L/L mice (WT: 0.33 pA; L/L: 0.24 pA). This difference could be attributed to a decrease in driving force since current-voltage analysis indicated that intracellular Na was increased in the L/L animals. Analysis of spontaneous channel noise indicated that the open probability was similar in the two genotypes(WT: 0.77; L/L: 0.80). Thus the increase in whole cell current is attributed to a difference in the density of conducting channels.


Assuntos
Aldosterona/farmacologia , Hipertensão/metabolismo , Rim/metabolismo , Canais de Sódio/metabolismo , Amilorida/farmacologia , Animais , Modelos Animais de Doenças , Diuréticos/farmacologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Hipertensão/genética , Rim/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Modelos Biológicos , Técnicas de Patch-Clamp , Canais de Sódio/genética , Sódio na Dieta/farmacocinética
19.
J Appl Physiol (1985) ; 93(5): 1881-7, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12381779

RESUMO

The amiloride-sensitive epithelial Na(+) channel (ENaC) is essential for fluid clearance from the airways. An experimental animal model with a reduced expression of ENaC, the alpha-ENaC transgenic rescue mouse, is prone to develop edema under hypoxia exposure. This strongly suggests an involvement of ENaC in the pathogenesis of pulmonary edema. To investigate the pathogenesis of this type of edema, primary cultures of tracheal cells from these mice were studied in vitro. An ~60% reduction in baseline amiloride-sensitive Na(+) transport was observed, but the pharmacological characteristics and physiological regulation of the channel were similar to those observed in cells from wild-type mice. Aprotinin, an inhibitor of serine proteases, blocked 50-60% of the basal transepithelial current, hypoxia induced downregulation of Na(+) transport, and beta-adrenergic stimulation was effective to stimulate Na(+) transport after the hypoxia-induced decrease. When downregulation of ENaC activity (such as observed under hypoxia) is added to a low "constitutive" ENaC expression, the resulting reduced Na(+) transport rate may be insufficient for airway fluid clearance and favor pulmonary edema.


Assuntos
Edema Pulmonar/metabolismo , Canais de Sódio/metabolismo , Sódio/metabolismo , Traqueia/metabolismo , Amilorida/farmacologia , Animais , Aprotinina/farmacologia , Transporte Biológico/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio , Hipóxia/metabolismo , Camundongos , Camundongos Transgênicos/genética , Valores de Referência , Inibidores de Serina Proteinase/farmacologia , Canais de Sódio/genética , Terbutalina/farmacologia , Traqueia/citologia
20.
Hum Mol Genet ; 11(23): 2829-36, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12393794

RESUMO

TMPRSS3 encodes a transmembrane serine protease that contains both LDLRA and SRCR domains and is mutated in non-syndromic autosomal recessive deafness (DFNB8/10). To study its function, we cloned the mouse ortholog which maps to Mmu17, which is structurally similar to the human gene and encodes a polypeptide with 88% identity to the human protein. RT-PCR and RNA in situ hybridization on rat and mouse cochlea revealed that Tmprss3 is expressed in the spiral ganglion, the cells supporting the organ of Corti and the stria vascularis. RT-PCR on mouse tissues showed expression in the thymus, stomach, testis and E19 embryos. Transient expression of wild-type or tagged TMPRSS3 protein showed a primary localization in the endoplasmic reticulum. The epithelial amiloride-sensitive sodium channel (ENaC), which is expressed in many sodium-reabsorbing tissues including the inner ear and is regulated by membrane-bound channel activating serine proteases (CAPs), is a potential substrate of TMPRSS3. In the Xenopus oocyte expression system, proteolytic processing of TMPRSS3 was associated with increased ENaC mediated currents. In contrast, 6 TMPRSS3 mutants (D103G, R109W, C194F, W251C, P404L, C407R) causing deafness and a mutant in the catalytic triad of TMPRSS3 (S401A), failed to undergo proteolytic cleavage and activate ENaC. These data indicate that important signaling pathways in the inner ear are controlled by proteolytic cleavage and suggest: (i) the existence of an auto-catalytic processing by which TMPRSS3 would become active, and (ii) that ENaC could be a substrate of TMPRSS3 in the inner ear.


Assuntos
Surdez/genética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Mutação de Sentido Incorreto/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo , Canais de Sódio/metabolismo , Animais , Sítios de Ligação , Western Blotting , Análise Mutacional de DNA , Primers do DNA/química , Surdez/metabolismo , Retículo Endoplasmático/metabolismo , Canais Epiteliais de Sódio , Feminino , Genes Recessivos/genética , Genótipo , Humanos , Hibridização In Situ , Técnicas In Vitro , Masculino , Camundongos , Oócitos/metabolismo , Órgão Espiral/metabolismo , Transporte Proteico , RNA Mensageiro/metabolismo , Coelhos , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Gânglio Espiral da Cóclea/metabolismo , Estria Vascular/metabolismo , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA