Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Curr Mol Med ; 4(4): 405-18, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15354871

RESUMO

Sphingolipids, historically described as potential reservoirs for bioactive lipids, presently define a new family of cellular mediators, joining the well-established glycerolipid-derived mediators of signal transduction such as diacylglycerol, phosphatidylinositides, and eicosanoids. Sphingolipid metabolism is clearly involved in the regulation of cell growth, differentiation, and programmed cell death. Indeed, a majority of the greater than four thousand studies conducted on sphingolipids during the past five years were investigations of the role of sphingolipids as cellular bioregulators. Studies spanning more than a decade have shown multiple interactions and intersections of the sphingolipid-mediated pathways and the eicosanoid pathway. This review will discuss the emerging mechanisms by which sphingolipids induce inflammatory responses via the eicosanoid pathway in addition to linking previous literature on sphingolipids and inflammation with newer findings of distinct roles for sphingosine-1-phosphate in regulating cyclooygenase-2 and ceramide-1-phosphate in the regulation of cytosolic phospholipase A2alpha. Finally, the relationship between bioactive sphingolipids and inflammation is discussed.


Assuntos
Inflamação/metabolismo , Esfingolipídeos/fisiologia , Ceramidas/metabolismo , Ciclo-Oxigenase 2 , Citocinas/metabolismo , Eicosanoides/metabolismo , Isoenzimas/metabolismo , Modelos Moleculares , Fosfolipases A/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , Transdução de Sinais , Esfingolipídeos/química , Esfingolipídeos/metabolismo , Esfingosina/análogos & derivados , Esfingosina/metabolismo
2.
J Biol Chem ; 276(48): 44848-55, 2001 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-11502750

RESUMO

The search for potential targets for ceramide action led to the identification of ceramide-activated protein phosphatases (CAPP). To date, two serine/threonine protein phosphatases, protein phosphatase 2A (PP2A) and protein phosphatase 1 (PP1), have been demonstrated to function as ceramide-activated protein phosphatases. In this study, we show that treatment with either anti-FAS IgM (CH-11) (150 ng/ml) or exogenous d-(e)-C(6-)ceramide (20 microm) induces the dephosphorylation of the PP1 substrates, serine/arginine-rich (SR) proteins, in Jurkat acute leukemia T-cells. The serine/threonine protein phosphatase inhibitor, calyculin A, but not the PP2A-specific inhibitor, okadaic acid, inhibited both FAS- and ceramide-induced dephosphorylation of SR proteins. Anti-FAS IgM treatment of Jurkat cells led to a significant increase in levels of endogenous ceramide beginning at 2 h with a maximal increase of 10-fold after 7 h. A 2-h pretreatment of Jurkat cells with fumonisin B(1) (100 microm), a specific inhibitor of CoA-dependent ceramide synthase, blocked 80% of the ceramide generated and completely inhibited the dephosphorylation of SR proteins in response to anti-FAS IgM. Moreover, pretreatment of Jurkat cells with myriocin, a specific inhibitor of serine-palmitoyl transferase (the first step in de novo synthesis of ceramide), also blocked FAS-induced SR protein dephosphorylation, thus demonstrating a role for de novo ceramide. These results were further supported using A549 lung adenocarcinoma cells treated with d-(e)-C(6-)ceramide. Dephosphorylation of SR proteins was inhibited by fumonisin B(1) and by overexpression of glucosylceramide synthase; again implicating endogenous ceramide generated de novo in regulating the dephosphorylation of SR proteins in response to FAS activation. These results establish a specific intracellular pathway involving both de novo ceramide generation and activation of PP1 to mediate the effects of FAS activation on SR proteins.


Assuntos
Ceramidas/biossíntese , Fumonisinas , Fosfoproteínas Fosfatases/metabolismo , Retículo Sarcoplasmático/metabolismo , Receptor fas/metabolismo , Western Blotting , Ácidos Carboxílicos/farmacologia , Linhagem Celular , Diacilglicerol Quinase/metabolismo , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Células Jurkat , Ácido Palmítico/metabolismo , Monoéster Fosfórico Hidrolases/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteína Fosfatase 2 , Serina/metabolismo , Esfingolipídeos/metabolismo , Treonina/metabolismo , Fatores de Tempo
3.
J Biol Chem ; 276(25): 22648-54, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11283022

RESUMO

Insulin regulates the inclusion of the exon encoding protein kinase C (PKC) betaII mRNA. In this report, we show that insulin regulates this exon inclusion (alternative splicing) via the phosphatidylinositol 3-kinase (PI 3-kinase) signaling pathway through the phosphorylation state of SRp40, a factor required for insulin-regulated splice site selection for PKCbetaII mRNA. By taking advantage of a well known inhibitor of PI 3-kinase, LY294002, we demonstrated that pretreatment of L6 myotubes with LY294002 blocked insulin-induced PKCbetaII exon inclusion as well as phosphorylation of SRp40. In the absence of LY294002, overexpression of SRp40 in L6 cells mimicked insulin-induced exon inclusion. When antisense oligonucleotides targeted to a putative SRp40-binding sequence in the betaII-betaI intron were transfected into L6 cells, insulin effects on splicing and glucose uptake were blocked. Taken together, these results demonstrate a role for SRp40 in insulin-mediated alternative splicing independent of changes in SRp40 concentration but dependent on serine phosphorylation of SRp40 via a PI 3-kinase signaling pathway. This switch in PKC isozyme expression is important for increases in the glucose transport effect of insulin. Significantly, insulin regulation of PKCbetaII exon inclusion occurred in the absence of cell growth and differentiation demonstrating that insulin-induced alternative splicing of PKCbetaII mRNA in L6 cells occurs in response to a metabolic change.


Assuntos
Processamento Alternativo/fisiologia , Insulina/fisiologia , Isoenzimas/genética , Músculo Esquelético/enzimologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteína Quinase C/genética , Animais , Sequência de Bases , Cicloeximida/farmacologia , Primers do DNA , Ativação Enzimática , Éxons , Músculo Esquelético/citologia , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Proteína Quinase C beta , Ratos
5.
Endocrinology ; 141(8): 2773-8, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10919262

RESUMO

Protein kinase C (PKCtheta) is a key enzyme in regulating a variety of cellular functions, including growth and differentiation. PKCtheta is the most abundant PKC isoform expressed in skeletal muscle; however, its role in differentiation and metabolism is not clear. We examined the effect of muscle cell differentiation on PKCtheta expression in human skeletal muscle cells from normal and type 2 diabetic subjects. Low levels of PKCtheta messenger RNA (mRNA) and protein were detected in human myoblasts from both types of subjects. Upon differentiation into myotubes, PKCtheta mRNA and protein were increased 12-fold in myotubes from normal subjects. In human skeletal muscle cells obtained from type 2 diabetic subjects, increases in PKCtheta mRNA and protein were not observed upon differentiation into myotubes although expression of other markers of differentiation and fusion increased. Cells from type 2 diabetic subjects also exhibited decreased insulin-stimulated glycogen synthase activity. To determine whether the up-regulation of PKCtheta was important for the metabolic actions of insulin, PKCtheta was overexpressed in L6 rat skeletal muscle cells. Increased expression of PKCtheta occurred with differentiation of skeletal muscle myoblasts to myotubes. Glycogen synthase activity was further increased in L6 myotubes stably transfected with the complementary DNA for PKCtheta. The decreased expression of PKCtheta found in cells from type 2 diabetic subjects may be linked to insulin resistance and decreased glycogen synthase activity.


Assuntos
Diferenciação Celular , Diabetes Mellitus Tipo 2/enzimologia , Glicogênio Sintase/metabolismo , Isoenzimas/genética , Músculo Esquelético/patologia , Proteína Quinase C/genética , Adulto , Animais , Linhagem Celular , Desoxiglucose/metabolismo , Diabetes Mellitus Tipo 2/patologia , Expressão Gênica , Humanos , Insulina/farmacologia , Pessoa de Meia-Idade , Músculo Esquelético/enzimologia , Proteína Quinase C/análise , Proteína Quinase C/fisiologia , RNA Mensageiro/análise , Ratos , Transfecção
6.
Am J Physiol Cell Physiol ; 279(3): C587-95, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10942708

RESUMO

Accelerated vascular smooth muscle cell (VSMC) proliferation contributes to the formation of atherosclerotic lesions. To investigate protein kinase C (PKC)-betaII functions with regard to glucose-induced VSMC proliferation, human VSMC from aorta (AoSMC), a clonal VSMC line of rat aorta (A10), and A10 cells overexpressing PKC-betaI (betaI-A10) and PKC-betaII (betaII-A10) were studied with the use of three techniques to evaluate glucose effects on aspects affecting proliferation. High glucose (25 mM) increased DNA synthesis and accelerated cell proliferation compared with normal glucose (5.5 mM) in AoSMC and A10 cells, but not in betaI-A10 and betaII-A10 cells. The PKC-betaII specific inhibitor CGP-53353 inhibited glucose-induced cell proliferation and DNA synthesis in AoSMC and A10 cells. In flow cytometry analysis, high glucose increased the percentage of A10 cells at 12 h after cell cycle initiation but did not increase the percentage of betaI-A10 or betaII-A10 cells entering S phase. PKC-betaII protein levels decreased before the peak of DNA synthesis, and high glucose further decreased PKC-betaII mRNA and protein levels in AoSMC and A10 cells. These results suggest that high glucose downregulates endogenous PKC-betaII, which then alters the normal inhibitory role of PKC-betaII in cell cycle progression, resulting in the stimulation of VSMC proliferation through acceleration of the cell cycle.


Assuntos
Glucose/farmacologia , Isoenzimas/metabolismo , Músculo Liso Vascular/citologia , Proteína Quinase C/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Divisão Celular/efeitos dos fármacos , Células Cultivadas , DNA/biossíntese , Regulação para Baixo , Inibidores Enzimáticos/farmacologia , Humanos , Músculo Liso Vascular/metabolismo , Ftalimidas/farmacologia , Proteína Quinase C beta , Ratos , Fase S/efeitos dos fármacos , Timidina/antagonistas & inibidores , Timidina/metabolismo , Fatores de Tempo
7.
Arch Biochem Biophys ; 372(1): 69-79, 1999 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-10562418

RESUMO

Insulin regulates a diverse array of signaling pathways involved in the control of growth, differentiation, proliferation, and metabolism. Insulin increases in glucose uptake via a protein kinase C-dependent pathway in target tissues such as fat and muscle are well documented. Insulin-regulated events, however, occur in all cells. The utilization of glucose as a preferred energy source is a ubiquitous event in eukaryotic cells. In NIH-3T3 fibroblasts, insulin treatment increased levels of the cPKC and nPKC activator, diacylglycerol. Insulin-responsive 2-[(3)H]deoxyglucose uptake was stimulated in a dose-dependent manner. The overexpression of protein kinase C (PKC)betaI, -betaII, -delta, -epsilon, and -zeta was used to investigate the specificity of PKC isozymes for insulin-sensitive glucose uptake. The stable overexpression of PKCbetaII, -delta, and -epsilon resulted in increases in insulin-stimulated 2-[(3)H]deoxyglucose uptake compared to vector control cells, while basal 2-deoxyglucose uptake levels were not elevated. Overexpression of PKCbetaI and PKCzeta isozymes had no further effect on basal or insulin-stimulated 2-deoxyglucose uptake. The PKC-specific inhibitor, CGP41251, blocked insulin effects on 2-deoxyglucose uptake but not its effects on tyrosine phosphorylation of cellular substrates. Insulin-stimulated 3-O-methylglucose uptake was also greater in cells overexpressing PKCbetaII, -delta, and -epsilon, compared to control cells. The increased responsiveness was not accompanied by conversion of 3T3 cells to the adipocyte phenotype or the increased expression of insulin receptors or glucose transporters (GLUT1-type). Insulin-stimulated recruitment of GLUT1 to plasma membranes of cells overexpressing PKCbetaII, -delta, and -epsilon, was greater than that in control cells. The data suggest that more than one PKC isozyme is involved in insulin signaling pathways in fibroblasts, resulting in increased GLUT1 transporter recruitment to cell membranes.


Assuntos
Glucose/metabolismo , Insulina/farmacologia , Isoenzimas/metabolismo , Proteína Quinase C/metabolismo , 3-O-Metilglucose/metabolismo , Células 3T3 , Animais , Transporte Biológico Ativo/efeitos dos fármacos , Desoxiglucose/metabolismo , Diglicerídeos/metabolismo , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Transportador de Glucose Tipo 1 , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Camundongos , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/genética , Receptor de Insulina/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia
8.
J Biol Chem ; 274(30): 21335-41, 1999 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-10409693

RESUMO

In the present study, we report that phosphatidic acid (PA) functions as a novel, potent, and selective inhibitor of protein phosphatase 1 (PP1). The catalytic subunit of PP1alpha was inhibited by PA dose-dependently in a noncompetitive manner with a K(i) value of 80 nM. The inhibition by PA was specific to PP1 as PA failed to inhibit protein phosphatase 2A (PP2A) or PP2B. Furthermore, PA was the most effective and potent inhibitor of PP1 compared with other phospholipids. Because we recently showed that ceramides activated PP1, we next examined the effects of PA on ceramide stimulation of PP1. PA inhibited both basal and ceramide-stimulated PP1 activities, and ceramide showed potent and stereoselective activation of PP1 in the presence of PA. Next, the effects of PA on ceramide-induced responses were examined. Molt-4 cells took up PA dose- and time-dependently such that by 1 and 3 h, uptake of PA was 0.37 and 0. 65% of total PA added, respectively. PA at 30 microM and calyculin A at 10 nM (an inhibitor of PP1 and PP2A at low concentrations), but not okadaic acid at 10 nM (a PP2A inhibitor at low concentrations) prevented poly(ADP-ribose) polymerase proteolysis induced by C(6)-ceramide. Moreover, the combination of PA with okadaic acid prevented retinoblastoma gene product dephosphorylation induced by C(6)-ceramide. These data suggest that PA functions as a specific regulator of PP1 and may reverse or counteract those effects of ceramide that are mediated by PP1, such as apoptosis and retinoblastoma gene product dephosphorylation.


Assuntos
Inibidores Enzimáticos/farmacologia , Ácidos Fosfatídicos/farmacologia , Fosfoproteínas Fosfatases/antagonistas & inibidores , Animais , Ceramidas/metabolismo , Ceramidas/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/metabolismo , Humanos , Células Jurkat , Ácidos Fosfatídicos/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteína Fosfatase 1 , Proteína Fosfatase 2 , Coelhos , Proteína do Retinoblastoma/metabolismo
9.
J Biol Chem ; 274(29): 20313-7, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10400653

RESUMO

The search for potential targets for ceramide action led to the identification of ceramide-activated protein phosphatases, which include protein phosphatase-2A (PP2A) and protein phosphatase-1 (PP1) with roles in regulating apoptosis and cell growth. Thus far, in vitro studies on ceramide-activated protein phosphatases have been restricted to the use of short chain ceramides, limiting the extent of mechanistic insight. In this study, we show that the long chain D-erythro-C18-ceramide activated PP2A (AB'C trimer), PP2Ac (catalytic subunit of PP2A), and PP1gammac and -alphac (catalytic subunits of PP1gamma and -1alpha isoforms, respectively) 2-6-fold in the presence of dodecane, a lipid-solubilizing agent, with 50% maximal activation achieved at approximately 10 microM D-erythro-C18-ceramide. The diastereoisomers of D-erythroC18-ceramide, D-threo-, and L-threo-C18-ceramide, as well as the enantiomeric L-erythro-C18-ceramide, did not activate PP1 or PP2A, but they inhibited PP1 and PP2A activity. The addition of phosphatidic acid decreased the basal activity of PP1c but also increased the stimulation by D-erythro-C18-ceramide from 1.8- to 2. 8-fold and decreased the EC50 of D-erythro-C18-ceramide to 4.45 microM. The addition of 150 mM KCl decreased the basal activity of PP1 and the dose of D-erythro-C18-ceramide necessary to activate PP1c (EC50 = 6.25 microM) and increased the ceramide responsiveness up to 10-17-fold. These studies disclose stereospecific activation of PP1 and PP2A by long chain natural ceramides under near physiologic ionic strengths in vitro. The implications of these studies for mechanisms of ceramide action are discussed.


Assuntos
Ceramidas/farmacologia , Ácidos Fosfatídicos/farmacologia , Fosfoproteínas Fosfatases/metabolismo , Cátions , Ativação Enzimática , Concentração Osmolar , Proteína Fosfatase 1 , Proteína Fosfatase 2
10.
FASEB J ; 13(1): 103-13, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9872935

RESUMO

Acute hyperglycemia may contribute to the progression of atherosclerosis by regulating protein kinase C (PKC) isozymes and by accelerating vascular smooth muscle cell (VSMC) proliferation. We investigated acute glucose regulation of PKCbeta gene expression in A10 cells, a rat aortic smooth muscle cell line. Western blot analysis showed that PKCbetaII protein levels decreased with high glucose (25 mM) compared to normal glucose (5.5 mM), whereas PKCbetaI levels were unaltered. PKCbeta mRNA levels were depleted by 60-75% in hyperglycemic conditions. To elucidate whether high glucose regulated PKCbeta expression via the common promoter for PKCbetaI and PKCbetaII, deletion constructs of the PKCbeta promoter ligated to CAT as reporter gene were transfected into A10 cells. Construct D (-411 to +179CAT) showed quenching in high glucose (25 mM) and suggested the involvement of a carbohydrate response element in the 5' promoter region of the PKCbeta gene. In actinomycin D-treated A10 cells, a 60% decrease in PKCbeta mRNA with high glucose treatment indicated that posttranscriptional destabilization by glucose was also occurring. We have demonstrated that glucose-induced posttranscriptional destabilization of PKCbetaII message is mediated via a nuclease activity present in the cytosol. The specificity of glucose to posttranscriptionally destabilize PKCbetaII mRNA, but not the PKCbetaI mRNA, was confirmed in both A10 cells and primary cultures from human aorta.


Assuntos
Regulação da Expressão Gênica , Glucose/metabolismo , Isoenzimas/genética , Músculo Liso Vascular/enzimologia , Proteína Quinase C/genética , RNA Mensageiro/metabolismo , Animais , Aorta/citologia , Linhagem Celular , Células Cultivadas , Meios de Cultura , Humanos , Hiperglicemia/enzimologia , Hiperglicemia/genética , Hiperglicemia/metabolismo , Isoenzimas/metabolismo , Músculo Liso Vascular/citologia , Regiões Promotoras Genéticas , Proteína Quinase C/metabolismo , Proteína Quinase C beta , Processamento Pós-Transcricional do RNA , Ratos , Transcrição Gênica
11.
Exp Cell Res ; 240(2): 349-58, 1998 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-9597008

RESUMO

The role of protein kinase C (PKC) on proliferation of A10 vascular smooth muscle cells (VSMC) was studied by overexpressing specific PKC-beta I and -beta II isozymes. PKC-beta I and -beta II are derived from alternative splicing of the exon encoding the carboxy-terminal (C-terminal) 50 or 52 amino acids, respectively. The differential functions of the two isozymes with regard to cell proliferation, DNA synthesis, and the cell cycle were investigated in A10 cells, a clonal cell line of VSMC from rat aorta, and in A10 cells overexpressing PKC-beta I and PKC-beta II (beta I-A10 and beta II-A10). PKC levels were increased three- to fourfold in heterogeneous cultures of stably transfected cells. Although doubling time of A10 cells was 36 h, the cell doubling time in beta I-A10 cells decreased by 12 h, and, in contrast, the doubling time of beta II-A10 cells increased by 12 h compared to A10 cells. The increase of [3H]thymidine (TdR) incorporation was accelerated and increased in beta I-A10 cells, but slowed and diminished in beta II-A10 cells compared to A10 and control cells transfected with empty vector. Cell cycle analysis of beta I-A10 cells showed an acceleration of S phase entry while beta II-A10 cells slowed S phase entry. These results suggest that PKC-beta I and PKC-beta II regulate cell proliferation bidirectionally and that PKC-beta I and PKC-beta II may have distinct and opposing functions as cell cycle check point mediators during late G1 phase and may regulate S phase entry in A10 VSMC.


Assuntos
Isoenzimas/fisiologia , Músculo Liso Vascular/citologia , Proteína Quinase C/fisiologia , Animais , Ciclo Celular , Divisão Celular , Expressão Gênica , Isoenzimas/genética , Proteína Quinase C/genética , Proteína Quinase C beta , Ratos
12.
J Biol Chem ; 273(2): 910-6, 1998 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-9422749

RESUMO

The protein kinase Cbeta (PKCbeta) gene encodes two isoforms, PKCbetaI and PKCbetaII, as a result of alternative splicing. The unique mechanism that underlies insulin-induced alternative splicing of PKCbeta pre-mRNA was examined in L6 myotubes. Mature PKCbetaII mRNA and protein rapidly increased >3-fold following acute insulin treatment, while PKCbetaI mRNA and protein levels remained unchanged. Mature PKCbetaII mRNA resulted from inclusion of the PKCbetaII-specific exon rather than from selection of an alternative polyadenylation site. Increased PKCbetaII expression was also not likely accounted for by transcriptional activation of the gene or increased stabilization of the PKCbetaII mRNA, and suggest that PKCbetaII expression is regulated primarily at the level of alternative splicing. Insulin effects on exon inclusion were observed as early as 15 min after insulin treatment; by 20 min, a new 5'-splice site variant of PKCbetaII was also observed. After 30 min, the longer 5'-splice site variant became the predominate species through activation of a downstream 5' splice site. Similar results were obtained using IGF-I. Although the role of this new PKCbetaII mRNA species is presently unknown, inclusion of either PKCbetaII-specific exon results in the same PKCbetaII protein.


Assuntos
Processamento Alternativo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Fator de Crescimento Insulin-Like I/farmacologia , Insulina/farmacologia , Isoenzimas/genética , Músculo Esquelético/efeitos dos fármacos , Proteína Quinase C/genética , Animais , Linhagem Celular , Éxons , Músculo Esquelético/citologia , Músculo Esquelético/enzimologia , Proteína Quinase C beta , RNA Mensageiro/genética , Ratos
13.
Mol Endocrinol ; 10(10): 1273-81, 1996 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9121494

RESUMO

Alternative splicing of pre-mRNA encoding the carboxy-terminal (C-terminal) exons of protein kinase C beta (PKC beta) leads to the expression of two protein isoforms, PKC beta 1 and PKC beta II, with the potential for different functions. PKC beta II expression is regulated by insulin via alternative mRNA splicing. A physiological consequence of its activation was investigated in L6 rat skeletal muscle cells expressing GLUT4 transporters, a cell line in which PKC is involved in glucose transport. We examined the contribution of PKC beta II for insulin-stimulated [3H]2-deoxyglucose uptake by constructing three PKC beta II C-terminal deletion mutants designated M216, M217, and M218. When transiently expressed in COS1 cells, M217, with nine amino acids deleted, demonstrated autophosphorylation activity 10-fold less than full-length PKC beta II. The mutants M218, with 13 amino acids deleted, and M216, with 52 amino acids deleted, demonstrated no autophosphorylation activity and are kinase negative. When transiently expressed in L6 myotubes, M217 inhibited insulin-stimulated 2-deoxyglucose uptake by 45% (with a 45% transfection efficiency) whereas M216 and M218, kinase-negative mutants, had no effect compared with cells transfected with control plasmid. Cotransfection of full-length PKC beta II with M217 was able to rescue the inhibition of insulin-stimulated 2-deoxyglucose uptake as compared with cotransfection of M217 with the control plasmid, suggesting that M217 acts as a dominant-negative. In contrast, cotransfection of full-length PKC beta I, the other alternatively spliced form, did not rescue inhibition of insulin-stimulated 2-deoxyglucose uptake by M217. To further demonstrate the involvement of PKC, specifically PKC beta II, in insulin-stimulated 2-deoxyglucose uptake, we used two inhibitors, CG41251 (a specific PKC inhibitor) and CG53353 (a PKC beta II-specific inhibitor at 1 microM). Both inhibited insulin-stimulated 2-deoxyglucose uptake 50-60% in L6 myotubes. We conclude that M217 may act as a specific PKC beta II dominant-negative and that PKC beta II is more specific for insulin-stimulated 2-deoxyglucose uptake in these cells than PKC beta I.


Assuntos
Desoxiglucose/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Músculo Esquelético/metabolismo , Proteína Quinase C/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Deleção de Genes , Dados de Sequência Molecular , Proteína Quinase C/genética , Ratos , Transdução de Sinais/genética
14.
Am J Physiol ; 270(2 Pt 2): F263-74, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8779886

RESUMO

Adenosine A1 receptor densities were increased in cultured LLC-PK1 and OK cells by chronic treatment with the adenosine receptor antagonists 1,3,7-trimethylxanthine (caffeine, 1 mM) and 1,3-dimethyl-8-cyclopentylxanthine [cyclopentyltheophylline (CPT), < or = 0.4 mM], respectively. The A1 receptor number per cell was increased twofold by 10-day treatments with 1 mM caffeine or 0.1 mM CPT, and the sodium-coupled glucose uptake was augmented twofold by 1 mM caffeine and sevenfold by 0.1 microM CPT (higher doses of CPT were progressively less stimulatory). Glucose uptake was blocked by acute (2-h) treatment with CPT, adenosine deaminase, or calphostin C. Caffeine (1 mM) or CPT (> or = 0.1 mM) inhibited cell proliferation for the first 10 days, then cell growth assumed a normal proliferative rate despite continued presence of antagonist. Cytosolic protein kinase C (PKC) beta-isoform immunoactivity and PKC-beta II mRNA were elevated at least twofold during 10 days of 0.1 mM CPT or 1 mM caffeine treatment. The sustained elevation in sodium-glucose symport and PKC activity observed with adenosine receptor antagonists was similar to acute (2-h) effects of the adenosine A1 agonist R(-)-N6-phenylisopropyladenosine (R-PIA, 0.1-1 microM). Moreover, cell proliferation was increased by adenosine (0.1 microM R-PIA), whereas Na-K-adenosinetriphosphatase activity was unaltered with chronic antagonist or acute adenosine treatments. Caffeine treatment may have some non-adenosine A1 receptor-mediated actions, because it slightly (30%) augmented protein kinase A activity. It is concluded that chronic exposure of proximal tubule cells to caffeine or CPT augments PKC and sodium-glucose transport but retards cell proliferation mainly via adenosine A1 receptor-mediated mechanisms.


Assuntos
Glucose/metabolismo , Rim/metabolismo , Proteína Quinase C/metabolismo , Receptores Purinérgicos P1/metabolismo , Regulação para Cima , Adenosina Desaminase/farmacologia , Animais , Transporte Biológico/fisiologia , Cafeína/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Linhagem Celular , Rim/citologia , Células LLC-PK1 , Fenilisopropiladenosina/farmacologia , Agonistas do Receptor Purinérgico P1 , Antagonistas de Receptores Purinérgicos P1 , Suínos , Teofilina/análogos & derivados , Teofilina/farmacologia
15.
Cell Growth Differ ; 6(11): 1353-65, 1995 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-8562473

RESUMO

Transmission electron microscopy was used to determine how immunogold labeling of PKC-alpha or -beta is modulated by the antitumor drug IFN (HuIFN alpha-2b) in the cytoplasm, membrane structures, and nucleus of rapidly dividing and confluent human glioma U-373 cells. Results showed that except for nuclear localization, there were no specific cytoplasmic organelles that PKC-alpha or -beta translocated to following HuIFN alpha-2b treatment. Electron micrographs of PKC-beta in proliferating cells depicted 1.34-fold more PKC-beta in the nucleus than in the cytoplasm and a 1-min HuIFN alpha-2b (500 units/ml) treatment transiently increased PKC-beta immunoreactivity in the cytoplasm (1.95-fold) and nucleus (1.97-fold). In confluent cells, incubation with HuIFN alpha-2b for 2 min significantly decreased cytoplasmic PKC-beta immunoreactivity by 37%, and no change was observed in nuclear PKC-beta labeling. PKC-alpha labeling in proliferating cells showed similar immunoreactivity in both control cytoplasm and nucleus. Treatment of proliferating cells with HuIFN alpha-2b for 2 min decreased PKC-alpha in the cytoplasm (59%) and nucleus (44%). In confluent cells, cytoplasmic PKC-alpha labeling decreased 59% at 1 min, 61% at 2 min, and 76% at 10 min of HuIFN alpha-2b treatment. Nuclear PKC-alpha decreased by 65% at 1 min, 80% at 2 min, and 62% at 10 min after HuIFN alpha-2b treatment. Western blots of total PKC-alpha in proliferating and confluent cells and PKC-beta in confluent cells showed similar results. However, Western blots of total PKC-alpha and -beta in proliferating cells did not demonstrate any significant changes in either PKC-alpha or -beta immunoreactivity following 1-min HuIFN alpha-2b treatment. These results suggest that treatment of proliferating U-373 cells with HuIFN alpha-2b for 1 min unfolds and exposes PKC-beta antigenic sites (hinge region) and increases in situ PKC-beta immunogold labeling.


Assuntos
Glioma/enzimologia , Interferon-alfa/farmacologia , Isoenzimas/genética , Proteína Quinase C/genética , Sequência de Aminoácidos , Especificidade de Anticorpos , Western Blotting , Imunofluorescência , Humanos , Imuno-Histoquímica , Isoenzimas/análise , Isoenzimas/imunologia , Microscopia Eletrônica , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteína Quinase C/análise , Proteína Quinase C/imunologia , Proteína Quinase C beta , Proteína Quinase C-alfa , RNA Mensageiro/análise , Sensibilidade e Especificidade , Células Tumorais Cultivadas/citologia , Células Tumorais Cultivadas/ultraestrutura
16.
J Biol Chem ; 270(22): 13326-32, 1995 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-7768933

RESUMO

Insulin regulates a diverse array of cellular signaling processes involved in the control of growth, differentiation, and cellular metabolism. Insulin increases glucose transport via a protein kinase C (PKC)-dependent pathway in BC3H-1 myocytes, but the function of specific PKC isozymes in insulin action has not been elucidated. Two isoforms of PKC beta result via alternative splicing of precursor mRNA. As now shown, both isoforms are present in BC3H-1 myocytes, and insulin induces alternative splicing of the PKC beta mRNA thereby switching expression from PKC beta I to PKC beta II mRNA. This effect occurs rapidly (15 min after insulin treatment) and is dose-dependent. The switch in mRNA is reflected by increases in the protein levels of PKC beta II. High levels of 12-0-tetradecanoylphorbol-13-acetate, which are commonly used to deplete or down-regulate PKC in cells, also induce the switch to PKC beta II mRNA following overnight treatment, and protein levels of PKC beta II reflected mRNA increases. To investigate the functional importance of the shift in PKC beta isoform expression, stable transfectants of NIH-3T3 fibroblasts overexpressing PKC beta I and PKC beta II were established. The overexpression of PKC beta II but not PKC beta I in NIH-3T3 cells significantly enhanced insulin effects on glucose transport. This suggests that PKC beta II may be more selective than PKC beta I for enhancing the glucose transport effects of insulin in at least certain cells and, furthermore, that insulin can regulate the expression of PKC beta II by alternative mRNA splicing.


Assuntos
Processamento Alternativo/efeitos dos fármacos , Insulina/farmacologia , Isoenzimas/genética , Proteína Quinase C/genética , Células 3T3 , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Isoenzimas/metabolismo , Camundongos , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Proteína Quinase C/metabolismo , Proteína Quinase C beta , RNA Mensageiro/genética , Acetato de Tetradecanoilforbol/farmacologia
17.
Biochem Biophys Res Commun ; 202(1): 562-70, 1994 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8037763

RESUMO

The presence of ANP prohormone immunoreactivity in rat GI tract suggests that it may be an extracardiac site of ANP synthesis. The aim of this study was to investigate the expression of ANP mRNA in the adult rat GI tract. ANP mRNA was detected by ribonuclease protection analysis in stomach, small and large intestines, and rectum/anus. The highest concentrations of ANP transcripts were found in the proximal stomach, antrum, proximal colon, and rectum/anus at levels that ranged from 1 to 10% of that found in cardiac ventricle. Northern blot analysis of total RNA from these tissues identified a single 0.9 kb ANP transcript similar to that detected in heart. Gel filtration chromatography of tissue extracts provided evidence for the presence of the complete ANP prohormone in proximal stomach, antrum, proximal colon and rectum/anus. These results demonstrate that the gene for ANP is expressed in specific regions of the rat GI tract, suggesting that tissue-specific differential regulation of ANP synthesis occurs within the GI tract.


Assuntos
Fator Natriurético Atrial/biossíntese , Sistema Digestório/metabolismo , Expressão Gênica , Animais , Fator Natriurético Atrial/isolamento & purificação , Northern Blotting , Cromatografia em Gel , Mucosa Gástrica/metabolismo , Intestino Delgado/metabolismo , Masculino , Miocárdio/metabolismo , Especificidade de Órgãos , Sondas RNA , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Ratos , Ratos Sprague-Dawley , Reto/metabolismo , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...