Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nephrol Dial Transplant ; 24(6): 1782-90, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19155535

RESUMO

BACKGROUND: Activation of protein kinase C (PKC) has been implicated in the pathogenesis of diabetic nephropathy where therapy targeting the beta isoform of this enzyme is in advanced clinical development. However, PKC-beta is also increased in various forms of human glomerulonephritis with several potentially nephrotoxic factors, other than high glucose, resulting in PKC-beta activation. Accordingly, we sought to examine the effects of PKC-beta inhibition in a non-diabetic model of progressive kidney disease. METHODS: Subtotally nephrectomized (STNx) rats were randomly assigned to receive either the selective PKC-beta inhibitor, ruboxistaurin or vehicle. In addition to functional and structural parameters, gene expression of the podocyte slit-pore diaphragm protein, nephrin, was also assessed. RESULTS: STNx animals developed hypertension, proteinuria and reduced glomerular filtration rate (GFR) in association with marked glomerulosclerosis and tubulointerstitial fibrosis. Glomerular nephrin expression was also reduced. Without affecting blood pressure, ruboxistaurin treatment attenuated the impairment in GFR and reduced the extent of both glomerulosclerosis and tubulointerstitial fibrosis in STNx rats. In contrast, neither proteinuria nor the reduction in nephrin expression was improved by ruboxistaurin. CONCLUSIONS: These findings indicate firstly that PKC-beta inhibition may provide a new therapeutic strategy in non-diabetic kidney disease and secondly that improvement in GFR is not inextricably linked to reduction in proteinuria.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Indóis/uso terapêutico , Nefropatias/tratamento farmacológico , Nefropatias/enzimologia , Maleimidas/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Animais , Modelos Animais de Doenças , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Rim/fisiopatologia , Nefropatias/patologia , Nefropatias/fisiopatologia , Masculino , Proteínas de Membrana/genética , Células Mesangiais/efeitos dos fármacos , Células Mesangiais/metabolismo , Proteína Quinase C beta , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta/farmacologia
2.
Am J Physiol Renal Physiol ; 295(6): F1705-14, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18815221

RESUMO

Conversion of normally quiescent mesangial cells into extracellular matrix-overproducing myofibroblasts in response to high ambient glucose and transforming growth factor (TGF)-beta(1) is central to the pathogenesis of diabetic nephropathy. Previously, we reported that mesangial cells respond to high glucose by generating reactive oxygen species (ROS) from NADPH oxidase dependent on protein kinase C (PKC) -zeta activation. We investigated the role of TGF-beta(1) in this action of high glucose on primary rat mesangial cells within 1-48 h. Both high glucose and exogenous TGF-beta(1) stimulated PKC-zeta kinase activity, as measured by an immune complex kinase assay and immunofluorescence confocal cellular imaging. In high glucose, Akt Ser473 phosphorylation appeared within 1 h and Smad2/3 nuclear translocation was prevented with neutralizing TGF-beta(1) antibodies. Neutralizing TGF-beta(1) antibodies, or a TGF-beta receptor kinase inhibitor (LY364947), or a phosphatidylinositol 3,4,5-trisphosphate (PI3) kinase inhibitor (wortmannin), prevented PKC-zeta activation by high glucose. TGF-beta(1) also stimulated cellular membrane translocation of PKC-alpha, -beta(1), -delta, and -epsilon, similar to high glucose. High glucose and TGF-beta(1) enhanced ROS generation by mesangial cell NADPH oxidase, as detected by 2,7-dichlorofluorescein immunofluorescence. This response was abrogated by neutralizing TGF-beta(1) antibodies, LY364947, or a specific PKC-zeta pseudosubstrate peptide inhibitor. Expression of constitutively active PKC-zeta in normal glucose caused upregulation of p22(phox), a likely mechanism of NADPH oxidase activation. We conclude that very early responses of mesangial cells to high glucose include autocrine TGF-beta(1) stimulation of PKC isozymes including PI3 kinase activation of PKC-zeta and consequent generation of ROS by NADPH oxidase.


Assuntos
Mesângio Glomerular/fisiologia , Hiperglicemia/fisiopatologia , NADPH Oxidases/metabolismo , Proteína Quinase C/metabolismo , Fator de Crescimento Transformador beta1/fisiologia , Animais , Primers do DNA , Ativação Enzimática , Mesângio Glomerular/fisiopatologia , Cinética , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo
3.
Proteins ; 72(1): 447-60, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18214957

RESUMO

Protein kinase C (PKC) isozymes are an important class of enzymes in cell signaling and as drug targets. They are involved in specific pathways and have selectivity towards certain ligands, despite their high sequence similarities. Ruboxistaurin is a specific inhibitor of PKC-beta. To understand the molecular determinants for the selectivity of ruboxistaurin, we derived the three-dimensional structures of the kinase domains of PKC-alpha, -betaI, and -zeta using homology modeling. Several binding orientations of ruboxistaurin in the binding sites of these PKC catalytic domains were analyzed, and a putative alternative binding site for PKC-zeta was identified in its kinase domain. The calculated free energy of binding correlates well with the IC(50) of the inhibitor against each PKC isozyme. A residue-based energy decomposition analysis attributed the binding free energy to several key residues in the catalytic sites of these enzymes, revealing potential protein-ligand interactions responsible for ligand binding. The contiguous binding site revealed in the catalytic domain of PKC-zeta provides avenues for selective drug design. The details of structural nuances for specific inhibition of PKC isozymes are presented in the context of the three-dimensional structures of this important class of enzymes.


Assuntos
Indóis/metabolismo , Maleimidas/metabolismo , Proteína Quinase C/metabolismo , Trifosfato de Adenosina/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Ligação de Hidrogênio , Indóis/química , Isoenzimas/química , Isoenzimas/metabolismo , Maleimidas/química , Modelos Moleculares , Dados de Sequência Molecular , Proteína Quinase C/química , Estrutura Terciária de Proteína , Alinhamento de Sequência , Homologia Estrutural de Proteína , Especificidade por Substrato , Termodinâmica
4.
Am J Physiol Endocrinol Metab ; 293(5): E1280-8, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17711990

RESUMO

Vascular endothelial growth factor (VEGF) is implicated in the development of proteinuria in diabetic nephropathy. High ambient glucose present in diabetes stimulates VEGF expression in several cell types, but the molecular mechanisms are incompletely understood. Here primary cultured rat mesangial cells served as a model to investigate the signal transduction pathways involved in high-glucose-induced VEGF expression. Exposure to high glucose (25 mM) significantly increased VEGF mRNA evaluated by real-time PCR by 3 h, VEGF cellular protein content assessed by immunoblotting or immunofluorescence within 24 h, and VEGF secretion by 24 h. High-glucose-induced VEGF expression was blocked by an antioxidant, Tempol, and antisense oligonucleotides directed against p22(phox), a NADPH oxidase subunit. Inhibition of protein kinase C (PKC)-beta(1) with the specific pharmacological inhibitor LY-333531 or inhibition of PKC-zeta with a cell permeable specific pseudosubstrate peptide also prevented enhanced VEGF expression in high glucose. Enhanced VEGF secretion in high glucose was prevented by Tempol, PKC-beta(1), or PKC-zeta inhibition. In normal glucose (5.6 mM), overexpression of p22(phox) or constitutively active PKC-zeta enhanced VEGF expression. Hypoxia inducible factor-1alpha protein was significantly increased in high glucose only by 24 h, suggesting a possible contribution to high-glucose-stimulated VEGF expression at later time points. Thus reactive oxygen species generated by NADPH oxidase, and both PKC-beta(1) and -zeta, play important roles in high-glucose-stimulated VEGF expression and secretion by mesangial cells.


Assuntos
Nefropatias Diabéticas/metabolismo , Glucose/administração & dosagem , Células Mesangiais/metabolismo , Chaperonas Moleculares/metabolismo , Proteína Quinase C/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Óxidos N-Cíclicos/farmacologia , Nefropatias Diabéticas/enzimologia , Imunofluorescência , Glucose/metabolismo , Indóis/farmacologia , Maleimidas/farmacologia , Chaperonas Moleculares/antagonistas & inibidores , NADPH Oxidases/antagonistas & inibidores , NADPH Oxidases/metabolismo , NADPH Oxidases/farmacologia , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C beta , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Marcadores de Spin , Fator A de Crescimento do Endotélio Vascular/genética
5.
Endocrinology ; 148(3): 1089-98, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17110421

RESUMO

Long-chain, monounsaturated fatty acids (FAs) stimulate secretion of the incretin hormone, glucagon-like peptide-1 (GLP-1) from the intestinal L cell. Because the atypical protein kinase C (PKC), PKCzeta, is involved in FA signaling in many cells, the role of PKCzeta in FA-induced GLP-1 secretion was investigated, using the murine GLUTag L cell line and primary rat intestinal L cells. GLUTag cells expressed mRNA for several PKC isoforms, including PKCzeta, and PKCzeta protein was localized throughout the cytoplasm in GLUTag and primary L cells as well as normal mouse and rat L cells. Treatment with oleic acid (150-1000 microm) for 2 h increased GLP-1 secretion (P < 0.001), and this was abrogated by the PKCzeta inhibitor ZI (P < 0.05) and PKCzeta small interfering RNA transfection (P < 0.05) but not inhibition of classical/novel PKC isoforms. Although most PKCzeta was localized in the particulate compartment of GLUTag cells, oleate treatment did not alter PKCzeta levels or activity in this cell fraction. GLUTag cells expressed mRNA for the Gq-coupled FA receptor GPR120; however, oleic acid did not induce any changes in Akt, MAPK, or calcium, and pretreatment with LY294002 and PD98059 to inhibit phosphatidylinositol 3-kinase and MAPK, respectively, did not prevent the effects of oleic acid. Finally, GLUTag cells also released GLP-1 in response to arachidonic acid (P < 0.001) but were not affected by other long-chain FAs. These findings demonstrate that PKCzeta is required for oleic acid-induced GLP-1 secretion. This enzyme may therefore serve as a therapeutic target to enhance GLP-1 release in type 2 diabetes.


Assuntos
Células Enteroendócrinas/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Ácido Oleico/farmacologia , Proteína Quinase C/fisiologia , Animais , Células Cultivadas , Embrião de Mamíferos , Células Enteroendócrinas/metabolismo , Camundongos , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Distribuição Tecidual
6.
Endocrinology ; 147(1): 222-31, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16365142

RESUMO

Metabolic flux through the hexosamine biosynthetic pathway (HBP) is increased in the presence of high glucose (HG) and potentially stimulates the expression of genes associated with the development of diabetic nephropathy. A number of synthetic processes are coupled to the HBP, including enzymatic intracellular O-glycosylation (O-GlcNAcylation), the addition of single O-linked N-acetylglucosamine monosaccharides to serine or threonine residues. Despite much data linking flow through the HBP and gene expression, the exact contribution of O-GlcNAcylation to HG-stimulated gene expression remains unclear. In glomerular mesangial cells, HG-stimulated plasminogen activator inhibitor-1 (PAI-1) gene expression requires the HBP and the transcription factor, Sp1. In this study, the specific role of O-GlcNAcylation in HG-induced PAI-1 expression was tested by limiting this modification with a dominant-negative O-linked N-acetylglucosamine transferase, by overexpression of neutral beta-N-acetylglucosaminidase, and by knockdown of O-linked beta-N-acetylglucosamine transferase expression by RNA interference. Decreasing O-GlcNAcylation by these means inhibited the ability of HG to increase endogenous PAI-1 mRNA and protein levels, the activity of a PAI-1 promoter-luciferase reporter gene, and Sp1 transcriptional activation. Conversely, treatment with the beta-N-acetylglucosaminidase inhibitor, O-(2-acetamido-2-deoxy-d-glucopyranosylidene)amino-N-phenylcarbamate, in the presence of normal glucose increased Sp1 O-GlcNAcylation and PAI-1 mRNA and protein levels. These findings demonstrate for the first time that among the pathways served by the HBP, O-GlcNAcylation, is obligatory for HG-induced PAI-1 gene expression and Sp1 transcriptional activation in mesangial cells.


Assuntos
Glucose/metabolismo , Células Mesangiais/fisiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Processamento de Proteína Pós-Traducional , Fator de Transcrição Sp1/metabolismo , Acetilglucosamina/metabolismo , Animais , Sequência de Bases , Regulação da Expressão Gênica , Glicosilação , Plasmídeos , Regiões Promotoras Genéticas , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Ativação Transcricional
7.
Kidney Int ; 68(6): 2526-41, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16316329

RESUMO

BACKGROUND: We postulated that in mesangial cells exposed to high glucose, protein kinase C-zeta (PKC-zeta) is necessary for the generation of reactive oxygen species (ROS) by nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and that the requirement of PKC-zeta for filamentous (F)-actin disassembly may involve ROS. To identify signaling mechanisms relevant to PKC-zeta activation and ROS generation, including phosphoinositide 3 kinase (PI3 kinase), we examined mesangial cell stimulation with platelet-derived growth factor (PDGF). METHODS: In primary rat mesangial cells cultured in 5.6 mmol/L or 30 mmol/L d-glucose, PKC-zeta expression was identified with immunoblotting and activity was analyzed in cell membrane immunoprecipitates and by confocal immunofluorescence imaging. ROS generation was measured by dichlorofluorescein fluorescence using confocal microscopy and was inhibited by transfection of antisense against NADPH subunits p22(phox) or p47(phox) or with Tempol. F-actin disassembly was observed by dual-channel confocal fluorescence imaging. PI3 kinase activity was detected by immunoblotting of phosphorylated Akt. RESULTS: In high glucose, generation of NADPH oxidase-dependent ROS was dependent on PKC-zeta. Conversely, sustained PKC-zeta activity was dependent on ROS generation, suggesting a positive feedback. PKC-zeta-dependent F-actin disassembly in high glucose required ROS generation. PDGF stimulated NADPH oxidase generation of ROS through a PKC-zeta mechanism that was independent of Akt phosphorylation and remained unchanged in high glucose. CONCLUSION: In high glucose, mesangial cell PKC-zeta is required for ROS generation from NADPH oxidase similar to PDGF stimulation of PKC-zeta-dependent ROS generation through a pathway independent of PI3 kinase. F-actin disassembly in high glucose also requires ROS. A positive feedback loop occurs between ROS and the activation of PKC-zeta in high glucose.


Assuntos
Glucose/farmacologia , Células Mesangiais/enzimologia , Proteína Quinase C/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Actinas/metabolismo , Animais , Células Cultivadas , Masculino , Proteínas de Membrana Transportadoras/genética , Células Mesangiais/citologia , Células Mesangiais/efeitos dos fármacos , NADPH Oxidases/genética , NADPH Oxidases/metabolismo , Oligonucleotídeos Antissenso , Fosfatidilinositol 3-Quinases/metabolismo , Fosfoproteínas/genética , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
8.
Exp Eye Res ; 80(5): 651-62, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15862172

RESUMO

Retinal pigment epithelial (RPE) cells express vascular endothelial growth factor (VEGF) in response to high glucose or hypoxia. We hypothesised that VEGF expression and secretion by RPE cells in high glucose and hypoxia are regulated by protein kinase C (PKC). Primary cultured RPE cells from Sprague-Dawley rats were growth-arrested for 48 hr in 0.5% FBS in 5.6 or 30 mm D-glucose. Cells were exposed to hypoxic conditions (<1% O(2), 5% CO(2)) for the last 15-18 hr of growth-arrest. PKC -alpha, -beta(1), -delta, -epsilon, and -zeta were expressed by RPE cells and exposure to high glucose for 48 hr had no effect on expression as demonstrated by Western immunoblotting. High glucose, hypoxia or VEGF stimulated translocation of a number of the PKC isozymes to the membrane or particulate fractions implying activation. In response to high glucose or acute phorbol myristate acetate (PMA) stimulation, VEGF mRNA analysed by RT-PCR was increased. Intracellular VEGF protein identified by immunoblotting and confocal immunofluorescence imaging was significantly increased by high glucose, hypoxia or acute PMA stimulation. Calphostin C or a specific inhibitor of PKC-zeta prevented high glucose-stimulated VEGF expression in high glucose. VEGF secretion, as measured by ELISA in the culture medium, was enhanced in hypoxia but not in high glucose. Following exposure of RPE cells to PMA for 24 hr, PKC-delta was significantly down regulated, whereas PKC-alpha, -beta, -epsilon and -zeta remained unchanged. Secretion of VEGF in normal or high glucose, or hypoxia was significantly reduced following treatment with PMA for 24 hr but not with the PKC-zeta inhibitor. We conclude that in high glucose and hypoxia PKC isozymes are activated and are necessary for VEGF expression. Secretion of VEGF is enhanced in hypoxia and appears to be regulated by PKC-delta. RPE cells may contribute to the pathogenesis of retinopathy caused by high glucose and hypoxia through the expression and secretion of VEGF that are regulated by PKC isozymes.


Assuntos
Glucose/farmacologia , Isoenzimas/metabolismo , Epitélio Pigmentado Ocular/química , Proteína Quinase C/metabolismo , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Western Blotting/métodos , Hipóxia Celular , Células Cultivadas , Ativação Enzimática , Ensaio de Imunoadsorção Enzimática/métodos , Isoenzimas/antagonistas & inibidores , Masculino , Microscopia Confocal , Naftalenos/farmacologia , Epitélio Pigmentado Ocular/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , RNA Mensageiro/análise , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Acetato de Tetradecanoilforbol/farmacologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
Curr Hypertens Rep ; 7(2): 148-54, 2005 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-15748541

RESUMO

High glucose activates a myriad of signaling and gene expression pathways in non-insulin-dependent target cells causing diabetes complications. One of the earliest responses to high glucose by vascular cells is the generation of reactive oxygen species (ROS) that act directly on intracellular proteins and DNA, or indirectly as second messengers, transforming these cells into disease phenotypes. ROS are produced by mitochondria and/or NADPH oxidase in all target cells exposed to high glucose studied to date. Reports using cell cultures and diabetic animal models indicate that inhibition of ROS generation prevents the amplification of signaling and gene expression that are implicated in vascular complications. These models convincingly demonstrate that maneuvers preventing ROS production attenuate or completely abrogate early micro- and macrovascular end-organ damage of diabetes, including nephropathy, retinopathy, and large-vessel atherosclerosis. Attention now turns to the development of more effective antioxidants that could be used in clinical trials in the prevention and treatment of diabetes complications.


Assuntos
Vasos Sanguíneos/patologia , Angiopatias Diabéticas/tratamento farmacológico , Indóis/uso terapêutico , Maleimidas/uso terapêutico , Proteína Quinase C/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Animais , Vasos Sanguíneos/metabolismo , Células Cultivadas , DNA/genética , Angiopatias Diabéticas/genética , Angiopatias Diabéticas/metabolismo , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/fisiologia , Humanos , Espaço Intracelular/metabolismo , Espaço Intracelular/ultraestrutura , Proteína Quinase C beta , Espécies Reativas de Oxigênio/antagonistas & inibidores , Resultado do Tratamento
10.
J Biol Chem ; 278(36): 33951-62, 2003 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-12821678

RESUMO

High glucose (HG) is the underlying factor contributing to long term complications of diabetes mellitus. The molecular mechanisms transforming the glomerular mesangial cell phenotype to cause nephropathy including diacylglycerol-sensitive protein kinase C (PKC) are still being defined. Reactive oxygen species (ROS) have been postulated as a unifying mechanism for HG-induced complications. We hypothesized that in HG an interaction between ROS generation, from NADPH oxidase, and PKC suppresses mesangial Ca2+ signaling in response to endothelin-1 (ET-1). In primary rat mesangial cells, growth-arrested (48 h) in 5.6 mM (NG) or 30 mm (HG) glucose, the total cell peak [Ca2+]i response to ET-1 (50 nM) was 630 +/- 102 nM in NG and was reduced to 159 +/- 15 nM in HG, measured by confocal imaging. Inhibition of PKC with phorbol ester down-regulation in HG normalized the ET-1-stimulated [Ca2+]i response to 541 +/- 74 nM. Conversely, an inhibitory peptide specific for PKC-zeta did not alter Ca2+ signaling in HG. Furthermore, overexpression of conventional PKC-beta or novel PKC-delta in NG diminished the [Ca2+]i response to ET-1, reflecting the condition observed in HG. Likewise, catalase or p47phox antisense oligonucleotide normalized the [Ca2+]i response to ET-1 in HG to 521 +/- 58 nM and 514 +/- 48 nM, respectively. Pretreatment with carbonyl cyanide m-chlorophenylhydrazone or rotenone did not restore Ca2+ signaling in HG. Detection of increased intracellular ROS in HG by dichlorofluorescein was inhibited by catalase, diphenyleneiodonium, or p47phox antisense oligonucleotide. HG increased p47phox mRNA by 1.7 +/- 0.1-fold as measured by reverse transcriptase-PCR. In NG, H2O2 increased membrane-enriched PKC-beta and -delta, suggesting activation of these isozymes. HG-enhanced immunoreactivity of PKC-delta visualized by confocal imaging was attenuated by diphenyleneiodium chloride. Thus, mesangial cell [Ca2+]i signaling in response to ET-1 in HG is attenuated through an interaction mechanism between NADPH oxidase ROS production and diacylglycerol-sensitive PKC.


Assuntos
Cálcio/metabolismo , Diglicerídeos/farmacologia , Endotelina-1/metabolismo , Glucose/metabolismo , NADPH Oxidases/metabolismo , Proteína Quinase C/química , Actinas/metabolismo , Animais , Catalase/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Diglicerídeos/metabolismo , Regulação para Baixo , Transporte de Elétrons , Proteínas de Fluorescência Verde , Peróxido de Hidrogênio/farmacologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Glomérulos Renais/citologia , Proteínas Luminescentes/metabolismo , Microscopia Confocal , Microscopia de Fluorescência , Mitocôndrias/metabolismo , Oligonucleotídeos/metabolismo , Oligonucleotídeos Antissenso/farmacologia , Oniocompostos/química , Peptídeos/química , Fosfoproteínas/metabolismo , Isoformas de Proteínas , Proteína Quinase C/metabolismo , Proteína Quinase C beta , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais , Fatores de Tempo , Transfecção
11.
Am J Physiol Renal Physiol ; 284(2): F303-12, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12388423

RESUMO

Endothelin-1 (ET-1) stimulates glomerular mesangial cell proliferation and extracellular matrix protein transcription through an ERK1/2-dependent pathway. In this study, we determined whether ET-1 activation of glomerular mesangial cell ERK1/2 is mediated through EGF receptor (EGF-R) transactivation and whether intact caveolae are required. We showed that ET-1 stimulated tyrosine phosphorylation of the EGF-R in primary cultured, growth-arrested rat mesangial cells. In response to ET-1, ERK1/2 phosphorylation was increased by 27 +/- 1-fold and attenuated by AG-1478, a specific EGF-R inhibitor, to 9 +/- 1-fold. Moreover, filipin III and beta-cyclodextrin, two cholesterol-depleting drugs known to disrupt caveolae, significantly reduced ET-1-induced phosphorylation of ERK1/2. In addition, preincubation of mesangial cells with a myristoylated peptide that binds to the caveolin-1 scaffolding domain diminished ET-1 activation of ERK1/2. ET-1 caused interaction of caveolin-1 with phosphorylated ERK1/2 identified by coimmunoprecipitation. Activation of ERK1/2 and its interaction with caveolin-1 were reduced by AG-1478, beta-cyclodextrin, or inhibition of PKC. Phosphorylated ERK1/2 localized at focal adhesion complexes along with phospho-caveolin-1, suggesting specific sites of compartmentalization of these signaling molecules. Hence, ET-1 activates mesangial cell ERK1/2 predominantly through a pathway involving EGF-R transactivation, leading to a mechanism involving attachment to caveolin-1, presumably in caveolae.


Assuntos
Caveolinas/fisiologia , Endotelina-1/fisiologia , Receptores ErbB/genética , Mesângio Glomerular/enzimologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Ativação Transcricional/fisiologia , Animais , Caveolina 1 , Células Cultivadas , Ativação Enzimática/fisiologia , Imunofluorescência , Mesângio Glomerular/citologia , Técnicas Imunológicas , Proteína Quinase 3 Ativada por Mitógeno , Fosforilação , Ratos , Ratos Sprague-Dawley
12.
J Biol Chem ; 277(37): 33833-41, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12105191

RESUMO

Increased flux through the hexosamine biosynthesis pathway (HBP) has been shown to stimulate the expression of a number of genes. We previously demonstrated in glomerular mesangial and endothelial cells that both high glucose concentrations and glucosamine activated the plasminogen activator inhibitor-1 (PAI-1) gene promoter through the transcription factor, Sp1; and that the glutamine:fructose-6-phosphate amidotransferase inhibitor, 6-diazo-5-oxonorleucine, inhibited the effect of high glucose, but not that of glucosamine. Here, we examined the role of protein kinase C (PKC) isoforms in the regulation of the PAI-1 promoter and Sp1 transcriptional activity by the HBP. In transient transfections, exposure to 2 mm glucosamine or 20 mm glucose for 4 days increased the activities of a PAI-1 promoter-luciferase reporter gene as well as the Sp1 transcriptional activation domain fused to the GAL4 DNA-binding domain cotransfected with a GAL4 promoter-luciferase reporter. Cotransfected dominant negative PKC-betaI and -delta completely blocked the induction of PAI-1 promoter transcription by both sugars, whereas only dominant negative PKC-betaI interfered with Sp1-GAL4 activation. Both glucosamine and high glucose stimulated the in vitro kinase activity of immunoprecipitated PKC-betaI and -delta. Furthermore, 6-diazo-5-oxonorleucine suppressed high glucose-induced PKC kinase activity and Sp1-GAL4 transcriptional activation. These findings demonstrate a requirement for the PKC-betaI and -delta signal transduction pathways in HBP-induced transcription.


Assuntos
Hexosaminas/biossíntese , Isoenzimas/fisiologia , Inibidor 1 de Ativador de Plasminogênio/genética , Regiões Promotoras Genéticas , Proteína Quinase C/fisiologia , Fator de Transcrição Sp1/genética , Ativação Transcricional , Animais , Células Cultivadas , Regulação da Expressão Gênica , Mesângio Glomerular/enzimologia , Glucosamina/farmacologia , Glucose/farmacologia , Glicosilação , Proteína Quinase C beta , Proteína Quinase C-delta , Ratos
13.
Am J Physiol Renal Physiol ; 282(6): F975-80, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11997313

RESUMO

High-glucose-induced activation of mesangial cell protein kinase C (PKC) contributes significantly to the pathogenesis of diabetic nephropathy. Excess glucose metabolism through the polyol pathway leads to de novo synthesis of both diacylglyerol (DAG) and phosphatidic acid, which may account for increased mesangial cell PKC-alpha, -beta, -delta, -epsilon, and -zeta activation/translocation observed within 48-h exposure to high glucose. Raised intracellular glucose causes generation of reactive oxygen species that may directly activate PKC isozymes and enhance their reactivity to vasoactive peptide signaling. In both diabetic rodent models of diabetes and cultured mesangial cells, PKC-beta appears to be the key isozyme required for the enhanced expression of transforming growth factor-beta(1), initiation of early accumulation of mesangial matrix protein, and increased microalbuminuria. Enhanced collagen IV expression by mesangial cells in response to vasoactive peptide hormone stimulation, e.g., endothelin-1, requires PKC-beta, -delta, -epsilon and -zeta. Loss of mesangial cell contractility to potent vasoactive peptides and coincident F-actin disassembly are due to high-glucose-activation of PKC-zeta. Inhibition of mesangial cell PKC isozyme activation in high glucose may prove to be the next important treatment for diabetic nephropathy.


Assuntos
Diabetes Mellitus/enzimologia , Mesângio Glomerular/enzimologia , Proteína Quinase C/metabolismo , Animais , Complicações do Diabetes , Nefropatias Diabéticas/enzimologia , Nefropatias Diabéticas/etiologia , Ativação Enzimática , Mesângio Glomerular/patologia , Humanos , Isoenzimas/metabolismo
14.
Am J Physiol Renal Physiol ; 282(1): F151-63, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11739123

RESUMO

In high glucose (HG), mesangial cells (MCs) lose their contractile response to endothelin-1 (ET-1) coincidently with filamentous (F)-actin disassembly. We postulated that these MC phenotypic changes are mediated by altered protein kinase C (PKC) isozyme activity, myosin light chain (MLC(20)) phosphorylation, or Ca(2+) signaling. MCs were growth arrested for 24 h in 0.5% fetal bovine serum (FBS)-DMEM in 5.6 (normal glucose; NG) or 30 mM glucose (high glucose; HG). In HG, the planar area was reduced [2,608 +/- 135 vs. 3,952 +/- 225 (SE) microm(2) in NG, P < 0.01, n = 31] with no contractile response to 0.1 microM ET-1. Mannitol did not affect cell size or ET-1 response. Confocal imaging of fluo 3- loaded cells revealed that the peak intensity of ET-1-induced Ca(2+) signaling was not altered in HG vs. NG. Immunoblotting of phosphorylated MLC(20) showed that HG increased mono- and decreased unphosphorylated MLC(20) (42 +/- 16 and 49 +/- 15 vs. 13 +/- 3 and 80 +/- 4% of total in NG, P < 0.05, n = 3), but the peak phosphorylation responses to ET-1 were identical in NG and HG. ET-1 stimulated translocation of PKC-delta and -epsilon from cytosolic to membrane and particulate fractions identically in NG and HG but did not cause PKC-zeta translocation. In HG, membrane accumulation of PKC-zeta was observed. Membrane PKC-zeta activity measured by immunoprecipitation and (32)P phosphorylation of PKC-epsilon pseudosubstrate peptide was 190 +/- 18% of NG (P < 0.01, n = 4), which was completely inhibited by pretreatment with a myristoylated peptide inhibitor (ZI). In HG, pretreatment with ZI for 24 h restored normal MC size and contractile and F-actin disassembly responses to ET-1. In conclusion, in HG, decreased MC size is due to decreased F-actin assembly, and loss of contractile response to ET-1 occurs in the presence of normal Ca(2+) signaling and normal MLC(20) phosphorylation. In HG, altered F-actin and contractile functions in MCs are mediated by PKC-zeta.


Assuntos
Actinas/metabolismo , Mesângio Glomerular/enzimologia , Glucose/farmacologia , Proteína Quinase C/metabolismo , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/fisiologia , Carcinógenos/farmacologia , Tamanho Celular , Células Cultivadas , Endotelina-1/farmacologia , Inibidores Enzimáticos/farmacologia , Mesângio Glomerular/citologia , Ionomicina/farmacologia , Ionóforos/farmacologia , Isoenzimas/metabolismo , Microscopia Confocal , Cadeias Leves de Miosina/metabolismo , Oligopeptídeos/farmacologia , Ésteres de Forbol/farmacologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-delta , Receptor de Endotelina A , Receptores de Endotelina/biossíntese , Serina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...