Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Biol Chem ; 276(33): 30871-7, 2001 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-11413133

RESUMO

Swelling of hepatocytes and other epithelia activates volume-sensitive ion channels that facilitate fluid and electrolyte efflux to restore cell volume, but the responsible signaling pathways are incompletely defined. Previous work in model HTC rat hepatoma cells has indicated that swelling elicits ATP release, which stimulates P2 receptors and activates Cl(-) channels, and that this mechanism is essential for hepatocellular volume recovery. Since P2 receptors are generally coupled to Ca(2+) signaling pathways, we determined whether hepatocellular swelling affected cytosolic [Ca(2+)], and if this involved a purinergic mechanism. Exposure of HTC cells to hypotonic media evoked an increase in cytosolic [Ca(2+)], which was followed by activation of K(+) and Cl(-) currents. Maneuvers that interfered with swelling-induced increases in cytosolic [Ca(2+)], including extracellular Ca(2+) removal and intracellular Ca(2+) store depletion with thapsigargin, inhibited activation of membrane currents and volume recovery. However, the swelling-induced increases in cytosolic [Ca(2+)] were unaffected by either extracellular ATP depletion with apyrase or blockade of P2 receptors with suramin. These findings indicate that swelling elicits an increase in hepatocellular Ca(2+), which is essential for ion channel activation and volume recovery, but that this increase does not stem from activation of volume-sensitive P2 receptors. Collectively, these observations imply that regulatory responses to hepatocellular swelling involve a dual requirement for a purinergic-independent Ca(2+) signaling cascade and a Ca(2+)-independent purinergic signaling pathway.


Assuntos
Trifosfato de Adenosina/fisiologia , Cálcio/fisiologia , Tamanho Celular , Hepatócitos/fisiologia , Animais , Canais de Cálcio/fisiologia , Receptores de Inositol 1,4,5-Trifosfato , Canais de Potássio/fisiologia , Ratos , Receptores Citoplasmáticos e Nucleares/fisiologia , Receptores Purinérgicos P2/fisiologia , Células Tumorais Cultivadas
2.
J Biol Chem ; 275(27): 20556-61, 2000 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-10783394

RESUMO

Despite abundant evidence for changes in mitochondrial membrane permeability in tumor necrosis factor (TNF)-mediated cell death, the role of plasma membrane ion channels in this process remains unclear. These studies examine the influence of TNF on ion channel opening and death in a model rat liver cell line (HTC). TNF (25 ng/ml) elicited a 2- and 5-fold increase in K(+) and Cl(-) currents, respectively, in HTC cells. These increases occurred within 5-10 min after TNF exposure and were inhibited either by K(+) or Cl(-) substitution or by K(+) channel blockers (Ba(2+), quinine, 0.1 mm each) or Cl(-) channel blockers (10 microm 5-nitro-2-(3-phenylpropylamino)benzoic acid and 0.1 mm N-phenylanthranilic acid), respectively. TNF-mediated increases in K(+) and Cl(-) currents were each inhibited by intracellular Ca(2+) chelation (5 mm EGTA), ATP depletion (4 units/ml apyrase), and the protein kinase C (PKC) inhibitors chelerythrine (10 micrometer) or PKC 19-36 peptide (1 micrometer). In contrast, currents were not attenuated by the calmodulin kinase II 281-309 peptide (10 micrometer), an inhibitor of calmodulin kinase II. In the presence of actinomycin D (1 micrometer), each of the above ion channel blockers significantly delayed the progression to TNF-mediated cell death. Collectively, these data suggest that activation of K(+) and Cl(-) channels is an early response to TNF signaling and that channel opening is Ca(2+)- and PKC-dependent. Our findings further suggest that K(+) and Cl(-) channels participate in pathways leading to TNF-mediated cell death and thus represent potential therapeutic targets to attenuate liver injury from TNF.


Assuntos
Apoptose/efeitos dos fármacos , Canais de Cloreto/metabolismo , Fígado/metabolismo , Canais de Potássio/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Alcaloides , Animais , Bário/farmacologia , Benzofenantridinas , Cálcio/farmacologia , Proteína Quinase Tipo 2 Dependente de Cálcio-Calmodulina , Proteínas Quinases Dependentes de Cálcio-Calmodulina/farmacologia , Canais de Cloreto/efeitos dos fármacos , Cloretos/metabolismo , Dactinomicina/farmacologia , Ácido Egtázico/farmacologia , Canais Iônicos/agonistas , Canais Iônicos/antagonistas & inibidores , Nitrobenzoatos/farmacologia , Técnicas de Patch-Clamp , Fenantridinas/farmacologia , Potássio/metabolismo , Canais de Potássio/efeitos dos fármacos , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Quinina , Ratos , Células Tumorais Cultivadas , ortoaminobenzoatos/farmacologia
3.
Am J Physiol Endocrinol Metab ; 278(2): E340-51, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10662719

RESUMO

To study effects of Bcl-x(L) in the pancreatic beta-cell, two transgenic lines were produced using different forms of the rat insulin promoter. Bcl-x(L) expression in beta-cells was increased 2- to 3-fold in founder (Fd) 1 and over 10-fold in Fd 2 compared with littermate controls. After exposure to thapsigargin (10 microM for 48 h), losses of cell viability in islets of Fd 1 and Fd 2 Bcl-x(L) transgenic mice were significantly lower than in islets of wild-type mice. Unexpectedly, severe glucose intolerance was observed in Fd 2 but not Fd 1 Bcl-x(L) mice. Pancreatic insulin content and islet morphology were not different from control in either transgenic line. However, Fd 2 Bcl-x(L) islets had impaired insulin secretory and intracellular free Ca(2+) ([Ca(2+)](i)) responses to glucose and KCl. Furthermore, insulin and [Ca(2+)](i) responses to pyruvate methyl ester (PME) were similarly reduced as glucose in Fd 2 Bcl-x(L) islets. Consistent with a mitochondrial defect, glucose oxidation, but not glycolysis, was significantly lower in Fd 2 Bcl-x(L) islets than in wild-type islets. Glucose-, PME-, and alpha-ketoisocaproate-induced hyperpolarization of mitochondrial membrane potential, NAD(P)H, and ATP production were also significantly reduced in Fd 2 Bcl-x(L) islets. Thus, although Bcl-x(L) promotes beta-cell survival, high levels of expression of Bcl-x(L) result in reduced glucose-induced insulin secretion and hyperglycemia due to a defect in mitochondrial nutrient metabolism and signaling for insulin secretion.


Assuntos
Apoptose , Expressão Gênica , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Mitocôndrias/fisiologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , Animais , Apoptose/efeitos dos fármacos , Glicemia/metabolismo , Cálcio/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Glucose/farmacologia , Teste de Tolerância a Glucose , Imuno-Histoquímica , Insulina/análise , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/ultraestrutura , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Cloreto de Potássio/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/análise , Ratos , Tapsigargina/farmacologia , Proteína bcl-X
4.
Metabolism ; 49(12): 1579-87, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11145120

RESUMO

This study investigates the mechanisms responsible for glucagon-like peptide-1 (GLP-1)-induced insulin secretion in Zucker diabetic fatty (ZDF) rats and their lean control (ZLC) littermates. Glucose, and 100 nmol/L GLP-1 (7-37 hydroxide) in the presence of stimulatory glucose concentrations, induced insulin secretion in islets from ZLC animals. In contrast, ZDF islets hypersecreted insulin at low glucose (5 mmol/L) and were poorly responsive to 15 mmol/L glucose stimulation, but increased insulin secretion following exposure to GLP-1. The insulin secretory response to 100 nmol/L GLP-1 was reduced by 88% in ZLC islets exposed to exendin 9-39. The intracellular Ca2+ concentration ([Ca2+]i) increased in fura-2-loaded ZLC islets following stimulation with 12 mmol/L glucose alone or GLP-1 in the presence of 12 mmol/L glucose. The increases in [Ca2+]i and insulin secretion in ZLC islets induced by GLP-1 were attenuated by 1 micromol/L nitrendipine. In contrast, neither glucose nor GLP-1 substantially increased [Ca2+]i in ZDF islets. Furthermore, insulin secretory responses to GLP-1 were not significantly inhibited in ZDF islets by nitrendipine. However, the insulin secretory response to GLP-1 in both ZLC and ZDF islets was ablated by cholera toxin. Our findings indicate that in ZLC islets, GLP-1 induces insulin secretion by a mechanism that depends on Ca2+ influx through voltage-dependent Ca2+ channels, whereas in ZDF islets, the action of GLP-1 is mediated by Ca2+-independent signaling pathways.


Assuntos
Cálcio/fisiologia , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus/metabolismo , Glucagon/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Obesidade , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Ratos Zucker/metabolismo , Animais , Peptídeo 1 Semelhante ao Glucagon , Secreção de Insulina , Ratos
5.
Diabetes ; 47(12): 1881-8, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9836519

RESUMO

The present study was undertaken to test the hypothesis that exposure to high glucose concentrations enhances insulin secretion in pancreatic islets from glucokinase-deficient mice. Insulin secretion and intracellular calcium ([Ca2+]i) were measured as the glucose concentration was increased from 2 to 26 mmol/l in islets from heterozygous glucokinase (GK)-deficient mice (GK+/-) and their wild-type littermates (GK+/+). Results obtained in islets incubated in 11.6 or 30 mmol/l glucose for 48-96 h were compared. GK+/- islets that had been incubated in 30 mmol/l glucose showed improved although not normal insulin secretory and [Ca2+]i responses to the standard glucose challenge as well as an enhanced ability to sense small amplitude glucose oscillations. These effects were associated with increased glucokinase activity and protein. In contrast, exposure of GK+/+ islets to 30 mmol/l glucose increased their basal insulin secretion but reduced their incremental secretory responses to glucose and their ability to detect small amplitude glucose oscillations. Thus exposure of GK+/- islets to 30 mmol/l glucose for 48-96 h enhanced their ability to sense and respond to a glucose stimulus, whereas similar exposure of GK+/+ islets induced evidence of beta-cell dysfunction. These findings provide a mechanistic framework for understanding why glucokinase diabetes results in mild hyperglycemia that tends not to increase over time. In addition, the absence of one allele of the glucokinase gene appears to protect against glucose-induced beta-cell dysfunction (glucose toxicity).


Assuntos
Glucoquinase/genética , Hiperglicemia/fisiopatologia , Insulina/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Cálcio/metabolismo , Relação Dose-Resposta a Droga , Glucoquinase/efeitos dos fármacos , Glucoquinase/metabolismo , Glucose/administração & dosagem , Glucose/farmacologia , Teste de Tolerância a Glucose , Secreção de Insulina , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Mutação
6.
J Biol Chem ; 273(38): 24457-64, 1998 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-9733737

RESUMO

Mutations in the hepatocyte nuclear factor-1alpha (HNF-1alpha) gene cause maturity onset diabetes of the young type 3, a form of type 2 diabetes mellitus. In mice lacking the HNF-1alpha gene, insulin secretion and intracellular calcium ([Ca2+]i) responses were impaired following stimulation with nutrient secretagogues such as glucose and glyceraldehyde but normal with non-nutrient stimuli such as potassium chloride. Patch clamp recordings revealed ATP-sensitive K+ currents (KATP) in beta-cells that were insensitive to suppression by glucose but normally sensitive to ATP. Exposure to mitochondrial substrates suppressed KATP, elevated [Ca2+]i, and corrected the insulin secretion defect. NAD(P)H responses to glucose were substantially reduced, and inhibitors of glycolytic NADH generation reproduced the mutant phenotype in normal islets. Flux of glucose through glycolysis in islets from mutant mice was reduced, as a result of which ATP generation in response to glucose was impaired. We conclude that hepatocyte nuclear factor-1alpha diabetes results from defective beta-cell glycolytic signaling, which is potentially correctable using substrates that bypass the defect.


Assuntos
Glicólise , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Proteínas Nucleares , Fatores de Transcrição/fisiologia , Trifosfato de Adenosina/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Ligação a DNA/fisiologia , Glucose/farmacologia , Glucose/fisiologia , Gliceraldeído/farmacologia , Fator 1 Nuclear de Hepatócito , Fator 1-alfa Nuclear de Hepatócito , Fator 1-beta Nuclear de Hepatócito , Técnicas In Vitro , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Canais de Potássio/fisiologia , Cloreto de Potássio/farmacologia , Transdução de Sinais , Tolbutamida/farmacologia , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética
7.
J Biol Chem ; 273(17): 10402-10, 1998 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-9553098

RESUMO

Although stimulation of insulin secretion by glucose is regulated by coupled oscillations of membrane potential and intracellular Ca2+ ([Ca2+]i), the membrane events regulating these oscillations are incompletely understood. In the presence of glucose and tetraethylammonium, transgenically derived beta-cells (betaTC3-neo) exhibit coupled voltage and [Ca2+]i oscillations strikingly similar to those observed in normal islets in response to glucose. Using these cells as a model system, we investigated the membrane conductance underlying these oscillations. Alterations in delayed rectifier or Ca2+-activated K+ channels were excluded as a source of the conductance oscillations, as they are completely blocked by tetraethylammonium. ATP-sensitive K+ channels were also excluded, since the ATP-sensitive K+ channel blocker tolbutamide substituted for glucose in inducing [Ca2+]i oscillations. Thapsigargin, which depletes intracellular Ca2+ stores, and maitotoxin, an activator of nonselective cation channels, both converted the glucose-dependent [Ca2+]i oscillations into a sustained elevation. On the other hand, both SKF 96365, a blocker of Ca2+ store-operated channels, and external Na+ removal suppressed the glucose-stimulated [Ca2+]i oscillations. Maitotoxin activated a nonselective cation current in betaTC3 cells that was attenuated by removal of extracellular Na+ and by SKF 96365, in the same manner to a current activated in mouse beta-cells following depletion of intracellular Ca2+ stores. Currents similar to these are produced by the mammalian trp-related channels, a gene family that includes Ca2+ store-operated channels and inositol 1,4,5-trisphosphate-activated channels. We found several of the trp family genes were expressed in betaTC3 cells by reverse transcriptase polymerase chain reaction using specific primers, but by Northern blot analysis, mtrp-4 was the predominant message expressed. We conclude that a conductance underlying glucose-stimulated oscillations in beta-cells is provided by a Ca2+ store depletion-activated nonselective cation current, which is plausibly encoded by homologs of trp genes.


Assuntos
Cálcio/metabolismo , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Glucose/farmacologia , Imidazóis/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Metotrexato/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais , Células Tumorais Cultivadas
8.
J Clin Invest ; 101(8): 1623-32, 1998 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-9541492

RESUMO

This study investigated the role of intracellular free Ca2+ concentration ([Ca2+]i) in apoptosis in MIN6 cells, an insulin secreting cell line, and in mouse islets. Thapsigargin, an inhibitor of sarcoendoplasmic reticulum Ca2+-ATPases (SERCA), caused a time- and concentration-dependent decrease in the viability of MIN6 cells and an increase in DNA fragmentation and nuclear chromatin staining changes characteristic of apoptosis. Two structurally distinct SERCA inhibitors, cyclopiazonic acid and 2,5-di-[t-butyl]-1,4-hydroquinone also caused apoptosis, but agents that increased [Ca2+]i by other mechanisms did not induce apoptosis in MIN6 cells. Carbachol- or ionomycin-releasible intracellular Ca2+ stores were completely depleted in cells treated by SERCA inhibitors, but not by other agents that increase [Ca2+]i. The ability of thapsigargin to induce cell death was not affected by blocking Ca2+ influx or by clamping [Ca2+]i with a cytosolic Ca2+ buffer suggesting that the process did not depend on changes in [Ca2+]i per se. However, application of the lipoxygenase inhibitors 5,8,11-eicosatrienoic acid and nordihydroguaiaretic acid partially prevented MIN6 cell apoptosis, while exposure of cells to the product of lipoxygenase, 12-hydroxy-[5,8,10,14]-eicosatetraenoic acid, caused apoptosis. In contrast, inhibition of cyclooxygenase with indomethacin did not abolish thapsigargin-induced apoptosis in MIN6 cells. Our findings indicate that thapsigargin causes apoptosis in MIN6 cells by depleting intracellular Ca2+ stores and leading to release of intermediate metabolites of arachidonic acid metabolism.


Assuntos
Apoptose/fisiologia , Ácido Araquidônico/metabolismo , Cálcio/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Animais , Apoptose/efeitos dos fármacos , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Linhagem Celular , Fragmentação do DNA/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Técnicas In Vitro , Secreção de Insulina , Líquido Intracelular/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Retículo Sarcoplasmático/enzimologia , Tapsigargina/farmacologia
9.
DNA Cell Biol ; 16(10): 1249-56, 1997 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9364936

RESUMO

Chemokines mediate their chemotactic, proinflammatory effects by binding to and activating a variety of specific receptors belonging to the G protein-coupled superfamily of seven-transmembrane serpentine receptors. We report the cloning, chromosomal localization, expression, and ligand binding of a novel CC chemokine receptor, CCR10. CCR10 is expressed primarily in placenta and fetal liver, and binds two of the CC chemokines, monocyte chemoattractant protein (MCP)-1 and MCP-3, with highest affinity. The KD for MCP-3 binding was 1 nM, and MCP-1 competed for MCP-3 binding with an IC50 of 1.2 nM. The CC chemokines MCP-4 and RANTES competed for MCP-3 binding with IC50 values of 7.5 and 5.4 nM, respectively. The chromosomal location of CCR10 was determined to coincide with the CC chemokine receptor cluster on chromosome 3 (3p21.31-3p21.32). These results indicate that CCR10 is a novel CC chemokine receptor with a unique expression pattern that would be consistent with a role in placental immunity or hematopoiesis.


Assuntos
Quimiocina CCL2/metabolismo , Citocinas , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas Quimioatraentes de Monócitos/metabolismo , Receptores de Quimiocinas/genética , Receptores de Quimiocinas/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Ligação Competitiva , Células COS , Quimiocina CCL5/metabolismo , Quimiocina CCL7 , Mapeamento Cromossômico , Cromossomos Humanos Par 3/genética , Clonagem Molecular , Feminino , Humanos , Cinética , Masculino , Dados de Sequência Molecular , Especificidade de Órgãos , RNA Mensageiro/análise , Receptores CCR10 , Homologia de Sequência de Aminoácidos
10.
J Biol Chem ; 271(50): 32241-6, 1996 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-8943282

RESUMO

Voltage-dependent delayed rectifier K+ channels regulate aspects of both stimulus-secretion and excitation-contraction coupling, but assigning specific roles to these channels has proved problematic. Using transgenically derived insulinoma cells (betaTC3-neo) and beta-cells purified from rodent pancreatic islets of Langerhans, we studied the expression and role of delayed rectifiers in glucose-stimulated insulin secretion. Using reverse-transcription polymerase chain reaction methods to amplify all known candidate delayed rectifier transcripts, the expression of the K+ channel gene Kv2.1 in betaTC3-neo insulinoma cells and purified rodent pancreatic beta-cells was detected and confirmed by immunoblotting in the insulinoma cells. betaTC3-neo cells were also found to express a related K+ channel, Kv3.2. Whole-cell patch clamp demonstrated the presence of delayed rectifier K+ currents inhibited by tetraethylammonium (TEA) and 4-aminopyridine, with similar Kd values to that of Kv2.1, correlating delayed rectifier gene expression with the K+ currents. The effect of these blockers on intracellular Ca2+ concentration ([Ca2+]i) was studied with fura-2 microspectrofluorimetry and imaging techniques. In the absence of glucose, exposure to TEA (1-20 mM) had minimal effects on betaTC3-neo or rodent islet [Ca2+]i, but in the presence of glucose, TEA activated large amplitude [Ca2+]i oscillations. In the insulinoma cells the TEA-induced [Ca2+]i oscillations were driven by synchronous oscillations in membrane potential, resulting in a 4-fold potentiation of insulin secretion. Activation of specific delayed rectifier K+ channels can therefore suppress stimulus-secretion coupling by damping oscillations in membrane potential and [Ca2+]i and thereby regulate secretion. These studies implicate previously uncharacterized beta-cell delayed rectifier K+ channels in the regulation of membrane repolarization, [Ca2+]i, and insulin secretion.


Assuntos
Ilhotas Pancreáticas/química , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/genética , 4-Aminopiridina/farmacologia , Animais , Sequência de Bases , Cálcio/metabolismo , Linhagem Celular , Canais de Potássio de Retificação Tardia , Citometria de Fluxo , Glucose/metabolismo , Potenciais da Membrana , Camundongos , Dados de Sequência Molecular , Canais de Potássio/química , Canais de Potássio/fisiologia , Ratos , Alinhamento de Sequência , Canais de Potássio Shab , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia
11.
Am J Physiol ; 270(1 Pt 1): E133-40, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8772485

RESUMO

Development of non-insulin-dependent diabetes mellitus (NIDDM) is associated with defects in glucose-stimulated insulin secretion. We have investigated Zucker diabetic fatty rats (ZDF), an animal model of NIDDM, and found that, compared with control islets, the expression of mRNA encoding C- and D-isoforms of alpha 1-subunits of beta-cell L-type voltage-dependent Ca2+ channels (VDCC) was significantly reduced in islets isolated from ZDF rats. This correlated with a substantial reduction of L-type Ca2+ currents (ICa) in ZDF beta-cells. Intracellular Ca2+ concentration responses in ZDF islets after glucose, KCI, or BAY K 8644 stimulation were markedly attenuated, whereas responses evoked by carbachol were unimpaired, consistent with a specific decrease in ICa in the diabetic islets. This reduction was accompanied by loss of pulsatile insulin secretion from ZDF islets treated with oscillatory increases of external glucose concentration. Our findings suggest that the attenuation of ICa in diabetic islets may contribute to the abnormal glucose-dependent insulin secretory responses associated with NIDDM and indicate that this defect is caused by decreased expression of genes encoding beta-cell VDCC alpha 1-subunits.


Assuntos
Canais de Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/metabolismo , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Cálcio/metabolismo , Canais de Cálcio/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Eletrofisiologia , Expressão Gênica , Glucose/farmacologia , Membranas Intracelulares/metabolismo , Ilhotas Pancreáticas/fisiopatologia , Masculino , Concentração Osmolar , Técnicas de Patch-Clamp , Cloreto de Potássio/farmacologia , RNA Mensageiro/metabolismo , Ratos , Ratos Zucker
13.
J Biol Chem ; 269(45): 27787-90, 1994 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-7961701

RESUMO

Glucose stimulation of pancreatic beta-cell insulin secretion is closely coupled to alterations in ion channel conductances and intracellular Ca2+ ([Ca2+]i). To further examine this relationship after augmentation of voltage-dependent K+ channel expression, transgenic mice were produced which specifically overexpress a human insulinoma-derived, tetraethylammonium (TEA)-insensitive delayed rectifier K+ channel in their pancreatic beta-cells as shown by immunoblot of isolated islets and immunohistochemical analysis of pancreas sections. Whole-cell current recordings confirmed the presence of high amplitude TEA-resistant K+ currents in transgenic islet cells, whose expression correlated with hyperglycemia and hypoinsulinemia. Stable overexpression of the channel in insulinoma cells attenuated glucose-activated increases in [Ca2+]i and prevented the induction of TEA-dependent [Ca2+]i oscillations. These results, employing the first ion channel transgenic mouse, demonstrate the importance of membrane potential regulation in excitation-secretion coupling in the pancreatic beta-cell.


Assuntos
Glucose/farmacologia , Ilhotas Pancreáticas/fisiologia , Canais de Potássio/biossíntese , Animais , Glicemia/metabolismo , Células CHO , Cálcio/metabolismo , Células Cultivadas , Cricetinae , Humanos , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Insulinoma/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neoplasias Pancreáticas/metabolismo , Canais de Potássio/isolamento & purificação , Canais de Potássio/fisiologia , Ratos , Tetraetilamônio , Compostos de Tetraetilamônio/farmacologia , Transfecção
14.
J Biol Chem ; 269(33): 21127-35, 1994 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-8063733

RESUMO

Rapid, transient induction of c-fos transcription follows treatment of cells with insulin and other growth factors. This is mediated through the serum response element (SRE), which binds the serum response factor (SRF), as well as accessory factors such as p62 ternary complex factor. Using a gel shift assay we found that formation of the ternary complex increased transiently within 2 min of insulin or phorbol ester treatment of several insulin-sensitive cell lines. However, mutations that prevented formation of this ternary complex did not inhibit insulin- or phorbol ester-stimulated induction of c-fos transcription in these cell lines. We also identified a novel SRF-containing multiprotein complex that forms on the SRE within 2 min following insulin, phorbol ester, or other growth factor treatment. Formation of this novel complex, called band 3, occurred rapidly and transiently, with a time course parallel to the induction of c-fos transcription. Band 3 also formed with gamma-actin and zif268/3 SRE probes. Methylation and carboxy ethylation interference analysis, as well as extensive SRE mutagenesis, suggest that only the SRF directly contacts the SRE in forming band 3. Formation of this novel complex appears to involve protein-protein interactions between SRF and other nuclear protein(s) that may play a role in growth factor stimulation of c-fos transcription.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Genes fos , Substâncias de Crescimento/farmacologia , Insulina/farmacologia , Proteínas Nucleares/metabolismo , Sequências Reguladoras de Ácido Nucleico , Células 3T3 , Animais , Sequência de Bases , DNA/metabolismo , Camundongos , Dados de Sequência Molecular , Ligação Proteica , Fator de Resposta Sérica , Acetato de Tetradecanoilforbol/farmacologia
15.
J Biol Chem ; 269(28): 18279-82, 1994 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-8034570

RESUMO

Non-insulin-dependent diabetes mellitus (NIDDM) is a metabolic disease associated with abnormal insulin secretion, the underlying mechanisms of which are unknown. Glucose-dependent signal transduction pathways were investigated in pancreatic islets derived from the db/db mouse, an animal model of NIDDM. After stimulation with glucose (4-12 mM), the changes in intracellular Ca2+ concentration ([Ca2+]i) were different; unlike control islets, db/db islets lacked an initial reduction of [Ca2+]i and the subsequent [Ca2+]i oscillations following stimulation with 12 mM glucose. The severity of these defects in Ca2+ signaling correlated with the age-dependent development of hyperglycemia. Similarly defective glucose-induced Ca2+ signaling were reproduced in control islets by pre-exposure to thapsigargin, a selective inhibitor of endoplasmic reticulum (ER) Ca(2+)-ATPase. Estimation of ATPase activities from rates of ATP hydrolysis and by immunoblot hybridization with an antiserum directed against the sarco/endoplasmic reticulum Ca(2+)-ATPase both demonstrated that the ER Ca(2+)-ATPase was almost entirely absent from db/db islets. The effects of inhibition of ER Ca(2+)-ATPase on insulin secretion were also examined; a 4-day exposure of control islets to 1 microM thapsigargin resulted in basal and glucose-stimulated insulin secretion levels similar to those found in db/db islets. These results suggest that aberrant ER Ca2+ sequestration underlies the impaired glucose responses in the db/db mouse and may play a role in defective insulin secretion associated with NIDDM.


Assuntos
Cálcio/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Retículo Endoplasmático/metabolismo , Glucose/farmacologia , Ilhotas Pancreáticas/metabolismo , Animais , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Calmodulina/antagonistas & inibidores , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/fisiopatologia , Retículo Endoplasmático/efeitos dos fármacos , Imidazóis/farmacologia , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Cinética , Camundongos , Camundongos Endogâmicos , Camundongos Mutantes , Valores de Referência , Terpenos/farmacologia , Tapsigargina
16.
Am J Physiol ; 266(6 Pt 1): E852-62, 1994 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-8023914

RESUMO

Stimulation of pancreatic islets of Langerhans with glucose results in changes in intracellular Ca2+ concentration ([Ca2+]i). With the use of mouse islets loaded with fura 2, the earliest glucose-induced alteration of [Ca2+]i was a pronounced decline in [Ca2+]i. This effect (phase 0) was evident 1 min after increasing extracellular glucose from 2 to 12 mM and was sustained for 3-5 min. Phase 0 was also observed when glucose was increased from 5 to 12 mM, indicating that it was not an experimental artifact resulting from substrate depletion. The [Ca2+]i-lowering effect of glucose was mimicked by D-glyceraldehyde but not by 2-deoxyglucose, pyruvate, glyburide, or 30 mM extracellular KCl. Mannoheptulose inhibited phase 0, whereas diazoxide, sodium azide, calmidazolium, or increasing extracellular [Ca2+] to 10 mM were all without effect. After the elevation of islet [Ca2+]i with 5 microM glyburide, 12 mM glucose caused a considerable transient decrease in [Ca2+]i. Under similar conditions, 5 mM caffeine attenuated phase 0, whereas 1 microM thapsigargin, a specific inhibitor of the sarcoplasmic and endoplasmic reticulum family of Ca(2+)-adenosinetriphosphatases (SERCA), almost completely inhibited any glucose-induced reduction of [Ca2+]i. These observations suggest that glucose causes an elevation of beta-cell SERCA activity triggered by factors generated during the cytosolic stages of glycolysis.


Assuntos
Glucose/farmacologia , Membranas Intracelulares/metabolismo , Ilhotas Pancreáticas/metabolismo , Terpenos/farmacologia , Animais , Cálcio/metabolismo , ATPases Transportadoras de Cálcio/antagonistas & inibidores , ATPases Transportadoras de Cálcio/metabolismo , Calmodulina/farmacologia , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Retículo Endoplasmático/metabolismo , Glucose/antagonistas & inibidores , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Retículo Sarcoplasmático/metabolismo , Tapsigargina
17.
J Biol Chem ; 269(20): 14359-62, 1994 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-8182038

RESUMO

Glucose stimulation of islet beta-cell insulin secretion is initiated by membrane depolarization and an elevation in intracellular free calcium concentration ([Ca2+]i) from a combination of influx through depolarization-activated Ca2+ channels and intracellular Ca2+ store release. Prevention of Ca2+ store refilling with thapsigargin produced a sustained depolarization, leading to enhanced Ca2+ influx and an elevation in [Ca2+]i in 12 mM glucose. Depletion of intracellular Ca2+ stores by external EGTA reduced [Ca2+]i and also caused a long-lasting depolarization. In single beta-cells, external EGTA activated an inward current, the voltage range and kinetic properties of which differed from those of voltage-dependent Ca2+ channels. A novel pathway thus exists in beta-cells by which depletion of endoplasmic reticulum Ca2+ stores results in the activation of an inward current that, by inducing depolarization, facilitates Ca2+ influx through voltage-gated Ca2+ channels. The physiological relevance of this pathway in the control of beta-cell function is indicated by the stimulation of insulin secretion by thapsigargin.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/fisiologia , Ilhotas Pancreáticas/fisiologia , Potenciais da Membrana , Animais , ATPases Transportadoras de Cálcio/antagonistas & inibidores , Glucose/farmacologia , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Terpenos/farmacologia , Tapsigargina , Fatores de Tempo
18.
J Biol Chem ; 269(15): 10979-82, 1994 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-8157622

RESUMO

An increase in cytosolic ATP following glucose metabolism by pancreatic beta-cells is the key signal initiating insulin secretion by causing blockade of ATP-dependent K+ channels (KATP). This induces membrane depolarization, leading to an elevation in cytosolic Ca2+ ([Ca2+]i) and insulin secretion. In this report we identify the critical metabolic step by which glucose initiates changes in beta-cell KATP channel activity, membrane potential, and [Ca2+]i. The signal stems from the glycolytic production of NADH during the oxidation of glyceraldehyde 3-phosphate, which is subsequently processed into ATP by mitochondria via the operation of discrete shuttle systems.


Assuntos
Cálcio/metabolismo , Glucose/farmacologia , Glicólise , Ilhotas Pancreáticas/fisiologia , NAD/metabolismo , Canais de Potássio/fisiologia , Transdução de Sinais , Trifosfato de Adenosina/metabolismo , Animais , Células Cultivadas , Citosol/metabolismo , Glucose/metabolismo , Gliceraldeído 3-Fosfato/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Niacinamida/farmacologia , Canais de Potássio/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
19.
J Biol Chem ; 268(14): 9953-6, 1993 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-8387528

RESUMO

Glucose-activated beta-cell insulin secretion depends upon elevation of intracellular calcium concentration, [Ca2+]i, which is thought to arise from Ca2+ influx through voltage-dependent calcium channels. Using fura-2-loaded mouse islets, we demonstrate, in fact, that the major component of the glucose-activated [Ca2+]i rise represents voltage-dependent intracellular Ca2+ release. Furthermore, the Ca2+ release pool possesses a novel pharmacology in that it is caffeine-sensitive but ryanodine-insensitive. In the absence of external Ca2+, glucose still caused intracellular Ca2+ release, an effect blockable by tetrodotoxin. However, depolarization of the islet with KCl in low Ca(2+)-containing solutions induced intracellular Ca2+ release, which was resistant to tetrodotoxin. We conclude that glucose release of intracellular Ca2+ is dependent upon depolarization alone, possibly through increasing inositol 1,4,5-trisphosphate production.


Assuntos
Cálcio/metabolismo , Glucose/farmacologia , Ilhotas Pancreáticas/fisiologia , Animais , Cafeína/farmacologia , Canais de Cálcio/fisiologia , Células Cultivadas , Ácido Egtázico/farmacologia , Fura-2 , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Cinética , Potenciais da Membrana/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Cloreto de Potássio/farmacologia , Rianodina/farmacologia , Tetrodotoxina/farmacologia
20.
J Biol Chem ; 266(13): 8576-82, 1991 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-1902475

RESUMO

Rapid stimulation of c-fos transcription by many agonists requires the serum response element (SRE), which binds at least two distinct nuclear proteins, p67SRF and p62TCF. Using nuclear protein extracts from 1321-N1 human astrocytoma cells, we investigated ligand-induced changes in binding of these proteins to SRE probes. In these cells c-fos mRNA expression can be induced by epidermal growth factor (EGF) through protein kinase C-independent pathways and by phorbol esters through protein kinase C. We detected two DNA-protein complexes that formed specifically with the SRE (bands 1 and 2). Band 2 formation was increased 4-6 min after stimulation with EGF as well as serum and phorbol esters; this peaked at 10-30 min and returned to basal levels by 60 min. Induction of band 2 formation preceded the onset and peak accumulation of c-fos mRNA (15 and 30 min after EGF stimulation, respectively) and its return to basal levels (by 1-2 h). Band 2 formation was also increased A431 cells stimulated with EGF and in HeLa and Swiss-3T3 cells stimulated with serum. We found that band 1 contained p67SRF bound to the SRE; band 2 contained p67SRF and a second protein. Gel shift analyses using [35S]methionine-labeled p67SRF and nonradioactive DNA probes suggested that hormone treatment most likely modified the second protein component of band 2. Transient transfection of 1321-N1 cells with plasmids containing point mutations that prevented band 2 formation in vitro also abolished induction of c-fos transcription in vivo as assayed by RNase protection analysis. Thus, hormone-stimulated formation of the protein-DNA complex represented by band 2 may be involved in the activation of c-fos transcription.


Assuntos
Elementos Facilitadores Genéticos , Fator de Crescimento Epidérmico/farmacologia , Mitógenos/farmacologia , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/genética , Proto-Oncogenes , Astrocitoma , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas Nucleares/genética , Testes de Precipitina , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-fos , Fator de Resposta Sérica , Transcrição Gênica , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA