Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Am J Physiol Cell Physiol ; 305(1): C111-20, 2013 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-23636454

RESUMO

Mutations in cystic fibrosis transmembrane regulator (CFTR), a chloride channel in the apical membranes of secretory epithelial cells, underlie the fatal genetic disorder cystic fibrosis. Certain CFTR mutations, including the common mutation ΔF508-CFTR, result in greatly decreased levels of active CFTR at the apical membrane. Direct interactions between CFTR and the cytoskeletal adaptors filamin-A (FlnA) and Na(+)/H(+) exchanger regulatory factor 1 (NHERF1) stabilize the expression and localization of CFTR at the plasma membrane. The scaffold protein receptor for activated C kinase 1 (RACK1) also stabilizes CFTR surface expression; however, RACK1 does not interact directly with CFTR and its mechanism of action is unknown. In the present study, we report that RACK1 interacts directly with FlnA in vitro and in a Calu-3 airway epithelial cell line. We mapped the interaction between RACK1 and FlnA to the WD4 and WD6 repeats of RACK1 and to a segment of the large rod domain of FlnA, consisting of immunoglobulin-like repeats 8-15. Disruption of the RACK1-FlnA interaction causes a reduction in CFTR surface levels. Our results suggest that a novel RACK1-FlnA interaction is an important regulator of CFTR surface localization.


Assuntos
Proteínas Contráteis/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas dos Microfilamentos/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores de Superfície Celular/metabolismo , Sequência de Aminoácidos , Linhagem Celular Tumoral , Proteínas Contráteis/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Filaminas , Proteínas de Ligação ao GTP/genética , Regulação da Expressão Gênica/fisiologia , Humanos , Proteínas dos Microfilamentos/genética , Dados de Sequência Molecular , Proteínas de Neoplasias/genética , Ligação Proteica , Transporte Proteico/fisiologia , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética
3.
Am J Physiol Cell Physiol ; 305(2): C133-46, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23515529

RESUMO

Mice deficient in Na-K-2Cl cotransporter (NKCC1) have been generated by targeted disruption of the gene encoding NKCC1 involving the carboxy terminus (CT-NKCC1) but not the amino terminus. We hypothesize that the resulting physiological defects are due to loss of proteins interacting with CT-NKCC1. Using a yeast two-hybrid approach, adaptor protein COMMD1 was found to bind to CT-NKCC1 (aa 1,040-1,212). Binding was verified in a yeast-independent system using GST-COMMD1 and myc-CT-NKCC1. Truncated COMMD1 and CT-NKCC1 peptides were used in binding assays to identify the site of interaction. The results demonstrate concentration-dependent binding of COMMD1 (aa 1-47) to CT-NKCC1 (aa 1,040-1,134). Endogenous COMMD1 was detected in pull downs using recombinant FLAG-CT-NKCC1; this co-pull down was blocked by COMMD1 (aa 1-47). CT-NKCC1 (aa 1,040-1,137) decreased basolateral membrane expression of NKCC1, and COMMD1 (aa 1-47) increased NKCC1 membrane expression. Downregulation of COMMD1 using silencing (si)RNA led to a transient loss of endogenous COMMD1 but did not affect activation of NKCC1 by hyperosmotic sucrose. Hyperosmolarity caused a transient increase in NKCC1 membrane expression, indicating regulated trafficking of NKCC1; downregulation of COMMD1 using siRNA reduced baseline (unstimulated) NKCC1 expression and blunted a transient elevation in NKCC1 membrane expression caused by hyperosmolarity. Constitutive downregulation of COMMD1 in HT29 engineered cells exhibited loss of COMMD1 and decreased NKCC1 membrane expression with no effect on activation of NKCC1. Loss of COMMD1 in Calu-3 cells and in HT29 cells led to reduced ubiquitinated NKCC1. The results indicate a role for COMMD1 in the regulation of NKCC1 membrane expression and ubiquitination.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Epiteliais/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Linhagem Celular , Clonagem Molecular , Cobre/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Transporte de Íons , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Simportadores de Cloreto de Sódio-Potássio/genética , Membro 2 da Família 12 de Carreador de Soluto , Técnicas do Sistema de Duplo-Híbrido
4.
Am J Physiol Renal Physiol ; 299(6): F1401-6, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20861079

RESUMO

Hypertonicity increases urea transport independently of, as well as synergistically with, vasopressin in the inner medullary collect duct (IMCD). We previously showed that hypertonicity does not increase the level of cAMP in the IMCD, but it does increase the level of intracellular calcium. Since we also showed that hypertonicity increases both the phosphorylation and biotinylation of the urea transporters UT-A1 and UT-A3, this would suggest involvement of a calcium-dependent protein kinase in the regulation of urea transport in the inner medulla. In this study, we investigated whether protein kinase C (PKC), which is present in the IMCD, is a regulator of urea permeability. We tested the effect of PKC inhibitors and activators on urea permeability in the isolated, perfused rat terminal IMCD. Increasing osmolality from 290 to 690 mosmol/kgH(2)O significantly stimulated (doubled) urea permeability; it returned to control levels on inhibition of PKC with either 10 µM chelerythrine or 50 µM rottlerin. To determine the potential synergy between vasopressin and PKC, phorbol dibutyrate (PDBu) was used to stimulate PKC. Vasopressin stimulated urea permeability 247%. Although PDBu alone did not change basal urea permeability, in the presence of vasopressin, it significantly increased urea permeability an additional 92%. The vasopressin and PDBu-stimulated urea permeability was reduced to AVP alone levels by inhibition of PKC. We conclude that hypertonicity stimulates urea transport through a PKC-mediated phosphorylation. Whether PKC directly phosphorylates UT-A1 and/or UT-A3 or phosphorylates it as a consequence of a cascade of activations remains to be determined.


Assuntos
Túbulos Renais Coletores/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteína Quinase C/metabolismo , Acetofenonas/farmacologia , Animais , Benzofenantridinas/farmacologia , Benzopiranos/farmacologia , Túbulos Renais Coletores/efeitos dos fármacos , Masculino , Concentração Osmolar , Permeabilidade , Dibutirato de 12,13-Forbol/farmacologia , Proteína Quinase C/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Ureia/metabolismo , Vasopressinas/farmacologia , Transportadores de Ureia
5.
Alcohol Clin Exp Res ; 34(9): 1659-69, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20586749

RESUMO

BACKGROUND: Rats chronically fed ethanol for 3 weeks presented a marked decreased in total hepatic Mg(2+) content and required approximately 12 days to restore Mg(2+) homeostasis upon ethanol withdrawal. This study was aimed at investigating the mechanisms responsible for the EtOH-induced delay. METHODS: Hepatocytes from rats fed ethanol for 3 weeks (Lieber-De Carli diet-chronic model), rats re-fed a control diet for varying periods of time following ethanol withdrawal, and age-matched control rats fed a liquid or a pellet diet were used. As acute models, hepatocytes from control animals or HepG2 cells were exposed to varying doses of ethanol in vitro for 8 minutes. RESULTS: Hepatocytes from ethanol-fed rats presented a marked inhibition of Mg(2+) accumulation and a defective translocation of PKCepsilon to the cell membrane. Upon ethanol withdrawal, 12 days were necessary for PKCepsilon translocation and Mg(2+) accumulation to return to normal levels. Exposure of control hepatocytes or HepG2 cells to a dose of ethanol as low as 0.01% for 8 minutes was already sufficient to inhibit Mg(2+) accumulation and PKCepsilon translocation for more than 60 minutes. Also in this model, recovery of Mg(2+) accumulation was associated with restoration of PKCepsilon translocation. The use of specific antisense in HepG2 cells confirmed the involvement of PKCepsilon in modulating Mg(2+) accumulation. CONCLUSIONS: Translocation of PKCepsilon isoform to the hepatocyte membrane is essential for Mg(2+) accumulation to occur. Both acute and chronic ethanol administrations inhibit Mg(2+) accumulation by specifically altering PKCepsilon translocation to the cell membrane.


Assuntos
Etanol/farmacologia , Hepatócitos/metabolismo , Magnésio/metabolismo , Proteína Quinase C-épsilon/metabolismo , Transporte Proteico/efeitos dos fármacos , Animais , Elementos Antissenso (Genética)/farmacologia , Técnicas de Cultura de Células , Células Cultivadas , Etanol/administração & dosagem , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Masculino , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
6.
J Biol Chem ; 285(22): 17166-76, 2010 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-20351101

RESUMO

Mutations in the chloride channel cystic fibrosis transmembrane regulator (CFTR) cause cystic fibrosis, a genetic disorder characterized by defects in CFTR biosynthesis, localization to the cell surface, or activation by regulatory factors. It was discovered recently that surface localization of CFTR is stabilized by an interaction between the CFTR N terminus and the multidomain cytoskeletal protein filamin. The details of the CFTR-filamin interaction, however, are unclear. Using x-ray crystallography, we show how the CFTR N terminus binds to immunoglobulin-like repeat 21 of filamin A (FlnA-Ig21). CFTR binds to beta-strands C and D of FlnA-Ig21 using backbone-backbone hydrogen bonds, a linchpin serine residue, and hydrophobic side-chain packing. We use NMR to determine that the CFTR N terminus also binds to several other immunoglobulin-like repeats from filamin A in vitro. Our structural data explain why the cystic fibrosis-causing S13F mutation disrupts CFTR-filamin interaction. We show that FlnA-Ig repeats transfected into cultured Calu-3 cells disrupt CFTR-filamin interaction and reduce surface levels of CFTR. Our findings suggest that filamin A stabilizes surface CFTR by anchoring it to the actin cytoskeleton through interactions with multiple filamin Ig repeats. Such an interaction mode may allow filamins to cluster multiple CFTR molecules and to promote colocalization of CFTR and other filamin-binding proteins in the apical plasma membrane of epithelial cells.


Assuntos
Proteínas Contráteis/química , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Imunoglobulinas/química , Proteínas dos Microfilamentos/química , Actinas/química , Sequência de Aminoácidos , Biotinilação , Membrana Celular/metabolismo , Biologia Computacional/métodos , Cristalografia por Raios X/métodos , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Filaminas , Humanos , Ligação de Hidrogênio , Espectroscopia de Ressonância Magnética , Dados de Sequência Molecular , Mutação , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos
7.
J Biol Chem ; 283(32): 22147-56, 2008 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-18550547

RESUMO

Airway epithelial Na-K-2Cl (NKCC1) cotransport is activated through hormonal stimulation and hyperosmotic stress via a protein kinase C (PKC) delta-mediated intracellular signaling pathway. Down-regulation of PKCdelta prevents activation of NKCC1 expressed in Calu-3 cells. Previous studies of this signaling pathway identified coimmunoprecipitation of PKCdelta with SPAK (Ste20-related proline alanine-rich kinase). We hypothesize that endogenous PKCdelta activates SPAK, which subsequently activates NKCC1 through phosphorylation. Double-stranded silencing RNA directed against SPAK reduced SPAK protein expression by 65.8% and prevented increased phosphorylation of NKCC1 and functional activation of NKCC1 during hyperosmotic stress, measured as bumetanide-sensitive basolateral to apical (86)Rb flux. Using recombinant proteins, we demonstrate direct binding of PKCdelta to SPAK, PKCdelta-mediated activation of SPAK, binding of SPAK to the amino terminus of NKCC1 (NT-NKCC1, amino acids 1-286), and competitive inhibition of SPAK-NKCC1 binding by a peptide encoding a SPAK binding site on NT-NKCC1. The carboxyl terminus of SPAK (amino acids 316-548) pulls down endogenous NKCC1 from Calu-3 total cell lysates and glutathione S-transferase-tagged NT-NKCC1 pulls down endogenous SPAK. In intact cells, hyperosmotic stress increased phosphorylated PKCdelta, indicating activation of PKCdelta, and activity of endogenous SPAK kinase. Inhibition of PKCdelta activity with rottlerin blocked the increase in SPAK kinase activity. The results indicate that PKCdelta acts upstream of SPAK to increase activity of NKCC1 during hyperosmotic stress.


Assuntos
Células Epiteliais/metabolismo , Pulmão/citologia , Proteína Quinase C-delta/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Linhagem Celular , Regulação para Baixo , Células Epiteliais/enzimologia , Pressão Osmótica , Fosforilação , Ligação Proteica , Proteínas Serina-Treonina Quinases/genética , Membro 2 da Família 12 de Carreador de Soluto
8.
Am J Physiol Cell Physiol ; 293(1): C294-304, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17409124

RESUMO

Previous studies from this laboratory demonstrated a role for protein kinase C (PKC)epsilon in the regulation of cAMP-dependent cystic fibrosis transmembrane regulator (CFTR) Cl channel function via binding of PKCepsilon to RACK1, a receptor for activated C kinase, and of RACK1 to human Na(+)/H(+) exchanger regulatory factor (NHERF1). In the present study, we investigated the role of RACK1 in regulating CFTR function in a Calu-3 airway epithelial cell line. Confocal microscopy and biotinylation of apical surface proteins demonstrate apical localization of RACK1 independent of actin. Mass spectrometric analysis of NHERF1 revealed copurification of tubulin, which, in in vitro binding assays, selectively binds to NHERF1, but not RACK1, via a PDZ1 domain. In binding and pulldown assays, we show direct binding of a PDZ2 domain to NHERF1, pulldown of endogenous NHERF1 by a PDZ2 domain, and inhibition of NHERF1-tubulin binding by a PDZ1 domain. Downregulation of RACK1 using double-stranded silencing RNA reduced the amount of RACK1 by 77.5% and apical expression of biotinylated CFTR by 87.4%. Expression of CFTR, NHERF1, and actin were not altered by treatment with siRACK1 or by nontargeting control silencing RNA, which, in addition, did not affect RACK1 expression. On the basis of these results, we model a RACK1 proteome consisting of PKCepsilon-RACK1-NHERF1-NHERF1-tubulin with a role in stable expression of CFTR in the apical plasma membrane of epithelial cells.


Assuntos
Membrana Celular/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C-épsilon/metabolismo , Receptores de Superfície Celular/metabolismo , Mucosa Respiratória/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Tubulina (Proteína)/metabolismo , Actinas/metabolismo , Biotinilação , Linhagem Celular , Polaridade Celular , Proteínas de Ligação ao GTP/genética , Humanos , Microscopia Confocal , Complexos Multiproteicos/metabolismo , Proteínas de Neoplasias/genética , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Mucosa Respiratória/citologia , Espectrometria de Massas em Tandem
9.
Biochemistry ; 45(34): 10270-7, 2006 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-16922502

RESUMO

Regulation of the CFTR Cl channel function involves a protein complex of activated protein kinase Cepsilon (PKCepsilon) bound to RACK1, a receptor for activated C kinase, and RACK1 bound to the human Na(+)/H(+) exchanger regulatory factor (NHERF1) in human airway epithelial cells. Binding of NHERF1 to RACK1 is mediated via a NHERF1-PDZ1 domain. The goal of this study was to identify the binding motif for human NHERF1 on RACK1. We examined the site of binding of NHERF1 on RACK1 using peptides encoding the seven WD40 repeat units of human RACK1. One WD repeat peptide, WD5, directly binds NHERF1 and the PDZ1 domain with similar EC(50) values, blocks binding of recombinant RACK1 and NHERF1, and pulls down endogenous RACK1 from Calu-3 cell lysate in a dose-dependent manner. The remaining WD repeat peptides did not block RACK1-NHERF1 binding. An 11-amino acid peptide encoding a site on the PDZ1 domain blocks binding of the WD5 repeat peptide with the PDZ1 domain. An N-terminal 12-amino acid segment of the WD5 repeat peptide, which comprises the first of four antiparallel beta-strands, dose-dependently binds to the PDZ1 domain of NHERF1 and blocks binding of the PDZ1 domain to RACK1. These results suggest that the binding site might form a beta-turn with topology sufficient for binding of NHERF1. Our results also demonstrate binding of NHERF to RACK1 at the WD5 repeat, which is distinct from the PKCepsilon binding site on the WD6 repeat of RACK1.


Assuntos
Células Epiteliais/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Receptores de Superfície Celular/metabolismo , Sistema Respiratório/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sítios de Ligação , Linhagem Celular , Sistema Livre de Células/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística , Células Epiteliais/citologia , Proteínas de Ligação ao GTP/genética , Humanos , Proteínas de Neoplasias/genética , Fosfoproteínas/genética , Ligação Proteica , Proteína Quinase C-épsilon/genética , Proteína Quinase C-épsilon/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Receptores de Quinase C Ativada , Receptores de Superfície Celular/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sistema Respiratório/citologia , Trocadores de Sódio-Hidrogênio/genética
10.
J Biol Chem ; 280(27): 25491-8, 2005 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-15899883

RESUMO

Activity of Na+-K+-2Cl- co-transport (NKCC1) in epithelia is thought to be highly regulated through phosphorylation and dephosphorylation of the transporter. Previous functional studies from this laboratory suggested a role for protein phosphatase 2A (PP2A) as a serine/threonine protein phosphatase involved in the regulation of mammalian tracheal epithelial NKCC1. We expand on these studies to characterize serine/threonine protein phosphatase(s) necessary for regulation of NKCC1 function and the interaction of the phosphatase(s) with proteins associated with NKCC1. NKCC1 activity was measured as bumetanide-sensitive 86Rb uptake or basolateral to apical 86Rb flux in primary cultures of human tracheal epithelial cells or in Calu-3 airway epithelial cells grown on Transwell filter inserts. Preincubation with 0.1 nm okadaic acid, a PP2A >> phosphatase 1 (PP1) inhibitor, increased NKCC1 activity 3.5-fold in human tracheal epithelial cells and 4.1-fold in Calu-3 cells. Calyculin, a PP1 >> PP2A inhibitor, did not alter NKCC1 activity or percent bumetanide-sensitive flux. The effect of OA was dose-dependent with an IC50 of 0.4 nm. The alpha1-adrenergic agonist methoxamine increased NKCC1 activity and transiently increased PP2A activity 3.8-fold but did not alter PP1 activity. OA augmented methoxamine-dependent stimulation of NKCC1 activity. PP1, PP2A, and PP2C but not PP2B were detected in lysates from Calu-3 cells by immunoblot analysis. PP1 was not detected in immunoprecipitates of NKCC1 and vice versa. PP2A co-immunoprecipitated with NKCC1 and protein kinase C-delta (PKC-delta) and was pulled down by a recombinant N terminus of NKCC1 consisting of amino acids 1-286. One novel finding is co-precipitation of STE20-related proline-alanine-rich kinase, a regulatory kinase for NKCC1, with PP2A and PKC-delta. The results suggest a model of actin serving as a scaffold for binding and association of PKC-delta, PP2A, and STE20-related proline-alanine-rich kinase. The role of the complex of serine/threonine protein kinases and a protein phosphatase is probably the maintenance of optimal phosphorylation of NKCC1 coincident with its physiological function in epithelial absorption and secretion.


Assuntos
Fosfoproteínas Fosfatases/metabolismo , Mucosa Respiratória/enzimologia , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Actinas/metabolismo , Agonistas alfa-Adrenérgicos/farmacologia , Transporte Biológico/fisiologia , Linhagem Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Humanos , Imunoprecipitação , Metoxamina/farmacologia , Ácido Okadáico/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Proteína Quinase C/metabolismo , Proteína Quinase C-delta , Proteína Fosfatase 1 , Proteína Fosfatase 2 , Proteínas Serina-Treonina Quinases/metabolismo , Mucosa Respiratória/citologia , Membro 2 da Família 12 de Carreador de Soluto , Traqueia/citologia
11.
Am J Physiol Cell Physiol ; 288(4): C906-12, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15590896

RESUMO

Direct binding of nonmuscle F-actin and the C2-like domain of PKC-delta (deltaC2-like domain) is involved in hormone-mediated activation of epithelial Na-K-2Cl cotransporter isoform 1 (NKCC1) in a Calu-3 airway epithelial cell line. The goal of this study was to determine the site of actin binding on the 123-amino acid deltaC2-like domain. Truncations of the deltaC2-like domain were made by restriction digestion and confirmed by nucleotide sequencing. His6-tagged peptides were expressed in bacteria, purified, and analyzed with a Coomassie blue stain for predicted size and either a 6xHis protein tag stain or an INDIA His6 probe for expression of the His6 tag. Truncated peptides were tested for competitive inhibition of binding of activated, recombinant PKC-delta with nonmuscle F-actin. Peptides from the NH2-terminal region, but not the COOH-terminal region, of the deltaC2-like domain blocked binding of activated PKC-delta to F-actin. The deltaC2-like domain and three NH2-terminal truncated peptides of 17, 83, or 108 amino acids blocked binding, with IC50 values ranging from 1.2 to 2.2 nmol (6-11 microM). NH2-terminal deltaC2-like peptides also prevented methoxamine-stimulated NKCC1 activation and pulled down endogenous actin from Calu-3 cells. The proximal NH2 terminus of the deltaC2-like domain encodes a beta1-sheet region. The amino acid sequence of the actin-binding domain is distinct from actin-binding domains in other PKC isotypes and actin-binding proteins. Our results indicate that F-actin likely binds to the beta1-sheet region of the deltaC2-like domain in airway epithelial cells.


Assuntos
Actinas/metabolismo , Proteína Quinase C/química , Proteína Quinase C/metabolismo , Mucosa Respiratória/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Ligação Competitiva/fisiologia , Linhagem Celular , Ativação Enzimática/fisiologia , Humanos , Dados de Sequência Molecular , Peptídeos/metabolismo , Ligação Proteica/fisiologia , Membro 2 da Família 12 de Carreador de Soluto
12.
Am J Physiol Cell Physiol ; 286(5): C1037-44, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15075202

RESUMO

In past studies, we demonstrated regulation of CFTR Cl channel function by protein kinase C (PKC)-epsilon through the binding of PKC-epsilon to RACK1 (a receptor for activated C-kinase) and of RACK1 to human Na(+)/H(+) exchanger regulatory factor (NHERF1). In this study, we investigated the site of RACK1 binding on NHERF1 using solid-phase and solution binding assays and pulldown, immunoprecipitation, and (36)Cl efflux experiments. Recombinant RACK1 binding to glutathione S-transferase (GST)-tagged PDZ1 domain of NHERF1 was 10-fold higher than its binding to GST-tagged PDZ2 domain of NHERF1. PDZ1 binds to RACK1 in a dose-dependent manner and vice versa, with similar binding constants of 1.67 and 1.26 microg, respectively. Interaction of the PDZ1 domain with RACK1 was not blocked by binding of activated PKC-epsilon to RACK1. A GST-tagged PDZ1 domain pulled down endogenous RACK1 from Calu-3 cell lysate. An internal 11-amino acid motif embedding the GYGF carboxylate binding loop of PDZ1 binds to RACK1, inhibits binding of recombinant NHERF1 and RACK1, pulls down endogenous RACK1 from Calu-3 cell lysate, and blocks coimmunoprecipitation of endogenous RACK1 with endogenous NHERF1 but does not affect cAMP-dependent activation of CFTR. A similar amino acid sequence in the PDZ2 domain did not bind RACK1. Our results indicate binding of Calu-3 RACK1 predominantly to the PDZ1 domain of NHERF1 at a site encompassing the GYGF loop of the PDZ1 domain and a site on RACK1 distinct from a PKC-epsilon binding site. CFTR activation by cAMP-generating agent is not affected by loss of RACK1-NHERF1 interaction.


Assuntos
Fosfoproteínas/química , Fosfoproteínas/fisiologia , Receptores de Superfície Celular/metabolismo , Mucosa Respiratória/metabolismo , Motivos de Aminoácidos/fisiologia , Linhagem Celular , Humanos , Proteína Quinase C/metabolismo , Proteína Quinase C-épsilon , Estrutura Terciária de Proteína/fisiologia , Receptores de Quinase C Ativada , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Mucosa Respiratória/citologia , Trocadores de Sódio-Hidrogênio
15.
Am J Physiol Cell Physiol ; 284(2): C487-96, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12388079

RESUMO

Activation of airway epithelial Na-K-2Cl cotransporter (NKCC)1 requires increased activity of protein kinase C (PKC)-delta, which localizes predominantly to the actin cytoskeleton. Prompted by reports of a role for actin in NKCC1 function, we studied a signaling mechanism linking NKCC1 and PKC. Stabilization of actin polymerization with jasplakinolide increased activity of NKCC1, whereas inhibition of actin polymerization with latrunculin B prevented hormonal activation of NKCC1. Protein-protein interactions among NKCC1, actin, and PKC-delta were verified by Western blot analysis of immunoprecipitated proteins. PKC-delta was detected in immunoprecipitates of NKCC1 and vice versa. Actin was also detected in immunoprecipitates of NKCC1 and PKC-delta. Pulldown of endogenous actin revealed the presence of NKCC1 and PKC-delta. Binding of recombinant PKC-delta to NKCC1 was not detected in overlay assays. Rather, activated PKC-delta bound to actin, and this interaction was prevented by a peptide encoding deltaC2, a C2-like domain based on the amino acid sequence of PKC-delta. deltaC2 also blocked stimulation of NKCC1 function by methoxamine. Immunofluorescence and confocal microscopy revealed PKC-delta in the cytosol and cell periphery. Merged images of cells stained for actin and PKC-delta indicated colocalization of PKC-delta and actin at the cell periphery. The results indicate that actin is critical for the activation of NKCC1 through a direct interaction with PKC-delta.


Assuntos
Citoesqueleto de Actina/metabolismo , Membrana Celular/metabolismo , Depsipeptídeos , Células Epiteliais/metabolismo , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Mucosa Respiratória/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Acetofenonas/farmacologia , Citoesqueleto de Actina/efeitos dos fármacos , Agonistas alfa-Adrenérgicos/farmacologia , Animais , Benzopiranos/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Bumetanida/farmacologia , Compartimento Celular/efeitos dos fármacos , Compartimento Celular/fisiologia , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Citosol/metabolismo , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Metoxamina/farmacologia , Peptídeos Cíclicos/farmacologia , Ligação Proteica/efeitos dos fármacos , Ligação Proteica/fisiologia , Proteína Quinase C-delta , Estrutura Terciária de Proteína/fisiologia , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Membro 2 da Família 12 de Carreador de Soluto , Tiazóis/farmacologia , Tiazolidinas
16.
J Biol Chem ; 277(25): 22925-33, 2002 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-11956211

RESUMO

Protein kinase C (PKC) regulation of cystic fibrosis transmembrane regulator (CFTR) chloride function has been demonstrated in several cell lines, including Calu-3 cells that express native, wild-type CFTR. We demonstrated previously that PKC epsilon was required for cAMP-dependent CFTR function. The goal of this study was to determine whether PKC epsilon interacts directly with CFTR. Using overlay assay, immunoprecipitation, pulldown and binding assays, we show that PKC epsilon does not bind to CFTR, but does bind to a receptor for activated C kinase (RACK1), a 37-kDa scaffold protein, and that RACK1 binds to Na(+)/H(+) exchange regulatory factor (NHERF1), a binding partner of CFTR. In vitro binding assays demonstrate dose-dependent binding of PKC epsilon to RACK1 which is inhibited by an 8-amino acid peptide based on the sequence of the sixth Trp-Asp repeat in RACK1 or by an 8-amino acid sequence in the V1 region of PKC epsilon, epsilon V1-2. A 4-amino acid sequence INAL (70-73) expressed in CFTR shares 50% homology to the RACK1 inhibitory peptide, but it does not bind PKC epsilon. NHERF1 and RACK1 bind in a dose-dependent manner. Immunofluorescence and confocal microscopy of RACK1 and CFTR revealed colocalization of the proteins to the apical and lateral regions of Calu-3 cells. The results indicate the RACK1 binds PKC epsilon and NHERF1, thus serving as a scaffold protein to anchor the enzyme in proximity to CFTR.


Assuntos
Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Isoenzimas/metabolismo , Proteínas de Neoplasias/metabolismo , Fosfoproteínas/metabolismo , Proteína Quinase C/metabolismo , Animais , Linhagem Celular , AMP Cíclico/metabolismo , Eletroforese em Gel de Poliacrilamida , Proteínas de Ligação ao GTP , Humanos , Immunoblotting , Insetos , Microscopia Confocal , Microscopia de Fluorescência , Oligonucleotídeos Antissenso/farmacologia , Peptídeos/química , Testes de Precipitina , Ligação Proteica , Proteína Quinase C-alfa , Proteína Quinase C-épsilon , Receptores de Quinase C Ativada , Receptores de Superfície Celular , Proteínas Recombinantes/metabolismo , Trocadores de Sódio-Hidrogênio
17.
Am J Physiol Lung Cell Mol Physiol ; 282(5): L1151-9, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11943682

RESUMO

In this study, we tested the hypothesis that intracellular Cl(-) (Cl) regulates the activity of protein kinase C (PKC)-delta and thus the activation of Na-K-Cl cotransport (NKCC1) in a Calu-3 cell line. The alpha(1)-adrenergic agonist methoxamine (MOX) and hypertonic sucrose increased Cl and increased or decreased intracellular volume, respectively, without changing Cl concentration ([Cl(-)](i)). Titration of [Cl(-)](i) from 20-140 mM in nystatin-permeabilized cell monolayers did not affect the baseline activity of PKC-delta, PKC-zeta, or rottlerin-sensitive NKCC1. At 200 mM Cl(-), rottlerin-sensitive NKCC1 was activated, and PKC isotypes were localized predominantly to a particulate fraction. MOX induced a biphasic increase in NKCC1 activity and PKC-delta in activity and particulate localization of PKC-delta and -zeta. Activity of NKCC1 and PKC-delta decreased with increasing Cl from 20 to 80 mM Cl then increased at 140-200 mM Cl apparently as an additive effect to high [Cl(-)](i) levels. Rottlerin inhibited the effects of MOX, which indicates that PKC-delta was required for activation of NKCC1. The results indicate that, in airway epithelial cells, a Cl electrochemical gradient alone is not sufficient to stimulate NKCC1 activity; rather, elevated activity of PKC-delta is necessary. Further, high Cl levels induce a subcellular redistribution of PKC-delta, which results in increased enzyme activity.


Assuntos
Cloretos/metabolismo , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , Mucosa Respiratória/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Acetofenonas/farmacologia , Agonistas alfa-Adrenérgicos/farmacologia , Benzopiranos/farmacologia , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Bumetanida/farmacologia , Células Cultivadas , Fibrose Cística/metabolismo , Diuréticos/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Isoenzimas/antagonistas & inibidores , Metoxamina/farmacologia , Pressão Osmótica , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Receptores Adrenérgicos alfa/metabolismo , Mucosa Respiratória/citologia , Membro 2 da Família 12 de Carreador de Soluto
18.
Biochim Biophys Acta ; 1589(1): 77-88, 2002 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-11909643

RESUMO

Hyperosmotic stress activates Na+-K+-2Cl- cotransport (NKCC1) in secretory epithelia of the airways. NKCC1 activation was studied as uptake of 36Cl or 86Rb in human tracheal epithelial cells (HTEC). Application of hypertonic sucrose or NaCl increased bumetanide-sensitive ion uptake but did not affect Na+/H+ and Cl-/OH-(HCO3-) exchange carriers. Hyperosmolarity decreased intracellular volume (Vi) after 10 min from 7.8 to 5.4 microl/mg protein and increased intracellular Cl- (Cl-i) from 353 to 532 nmol/mg protein. Treatment with an alpha-adrenergic agent rapidly increased Cl-i and Vi in a bumetanide-sensitive manner, indicating uptake of ions by NKCC1 followed by osmotically obligated water. These results indicate that HTEC act as osmometers but lose intracellular water slowly. Hyperosmotic stress also increased the activity of PKC-delta and of the extracellular signal-regulated kinase ERK subgroup of the MAPK family. Activity of stress-activated protein kinase JNK was not affected by hyperosmolarity. PD-98059, an inhibitor of the ERK cascade, reduced ERK activity and bumetanide-sensitive 36Cl uptake. PKC inhibitors blocked activation of ERK indicating that PKC may be a downstream activator of ERK. The results indicate that hyperosmotic stress activates NKCC1 and this activation is regulated by PKC-delta and ERK.


Assuntos
Amilorida/análogos & derivados , Proteína Quinase C/metabolismo , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Traqueia/metabolismo , Ácido 4,4'-Di-Isotiocianoestilbeno-2,2'-Dissulfônico/farmacologia , Amilorida/farmacologia , Bumetanida/farmacologia , Células Cultivadas , Cloro/análise , Cloro/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Células Epiteliais/metabolismo , Flavonoides/farmacologia , Humanos , Proteínas Quinases Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Pressão Osmótica , Rubídio/metabolismo , Cloreto de Sódio , Membro 2 da Família 12 de Carreador de Soluto , Sacarose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...