Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 199
Filtrar
1.
Diabetes Obes Metab ; 14(11): 1010-9, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22687049

RESUMO

AIMS: Induction of inducible nitric oxide synthase (iNOS) in pancreatic islets leads to exaggerated nitric oxide (NO) production associated with dysfunctional ß-cells. We examined insulin secretion, iNOS expression and its relationship to the cAMP system in islets from human type 2 diabetes. METHODS: Insulin, glucagon and cAMP were analysed by RIA; iNOS or phosphodiesterase (PDE) expression by quantitative PCR (qPCR), Western blot and confocal microscopy; cell viability by MTS. RESULTS: Diabetic islets displayed impaired insulin and glucagon responses to glucose, disturbed cAMP generation and high inducible nitric oxide synthase (iNOS) mRNA and protein expression. Confocal microscopy showed iNOS protein expression in diabetic islets being confined to insulin, glucagon and somatostatin cells. Culture of diabetic islets at 5.5 mmol/l glucose with dibutyryl-cAMP (Bt(2) -cAMP) for 24 h was accompanied by marked suppression of iNOS mRNA, reduced nitrite production and increased insulin secretion. Diabetic islets displayed marked increase in PDE3A and PDE3B mRNA expression. Short-time incubation of diabetic islets showed, among the PDE inhibitors tested, cilostazol being most favourable to increase insulin secretion. Diabetic islets were most susceptible to long-term (72 h) culture at high glucose (20 mmol/l) reacting with increased apoptosis. Bt(2) -cAMP and the PDE inhibitors cilostazol, milrinone and IBMX efficiently increased cell viability at high glucose during culture. Defective glucose-stimulated insulin release upon induction of iNOS was restored by iNOS inhibitor aminoguanidine. CONCLUSION: Our results suggest that in islets from type 2 diabetes, stimulatory effects in certain cAMP-compartments induced by PDE inhibitors might play a central role in the suppression of iNOS, resulting in increased ß-cell viability and improved secretory response to glucose.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Glucagon/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Animais , Western Blotting , Diabetes Mellitus Tipo 2/imunologia , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Microscopia Confocal , Óxido Nítrico/metabolismo , Ratos
2.
Diabetologia ; 53(9): 1927-34, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20526761

RESUMO

AIMS/HYPOTHESES: To investigate the effects of extracellular purines on insulin secretion from mouse pancreatic islets. METHODS: Mouse islets and beta cells were isolated and examined with mRNA real-time quantification, cAMP quantification and insulin and glucagon secretion. ATP release was measured in MIN6c4 cells. Insulin and glucagon secretion were measured in vivo after glucose injection. RESULTS: Enzymatic removal of extracellular ATP at low glucose levels increased the secretion of both insulin and glucagon, while at high glucose levels insulin secretion was reduced and glucagon secretion was stimulated, indicating an autocrine effect of purines. In MIN6c4 cells it was shown that glucose does induce release of ATP into the extracellular space. Quantitative real-time PCR demonstrated the expression of the ADP receptors P2Y(1) and P2Y(13) in both intact mouse pancreatic islets and isolated beta cells. The stable ADP analogue 2-MeSADP had no effect on insulin secretion. However, co-incubation with the P2Y(1) antagonist MRS2179 inhibited insulin secretion, while co-incubation with the P2Y(13) antagonist MRS2211 stimulated insulin secretion, indicating that ADP acting via P2Y(1) stimulates insulin secretion, while signalling via P2Y(13) inhibits the secretion of insulin. P2Y(13) antagonism through MRS2211 per se increased the secretion of both insulin and glucagon at intermediate (8.3 mmol/l) and high (20 mmol/l) glucose levels, confirming an autocrine role for ADP. Administration of MRS2211 during glucose injection in vivo resulted in both increased secretion of insulin and reduced glucose levels. CONCLUSIONS/INTERPRETATION: In conclusion, ADP acting on the P2Y(13) receptors inhibits insulin release. An antagonist to P2Y(13) increases insulin release and could be evaluated for the treatment of diabetes.


Assuntos
Difosfato de Adenosina/metabolismo , Insulina/metabolismo , Receptores Purinérgicos P2/metabolismo , Difosfato de Adenosina/análogos & derivados , Difosfato de Adenosina/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Apirase/metabolismo , Linhagem Celular , Células Cultivadas , AMP Cíclico/metabolismo , Feminino , Glucagon/metabolismo , Técnicas In Vitro , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Reação em Cadeia da Polimerase , Receptores Purinérgicos P2Y1 , Tionucleotídeos/farmacologia
3.
Acta Physiol (Oxf) ; 195(3): 375-83, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18764864

RESUMO

AIM: The role of islet nitric oxide (NO) production in insulin-releasing mechanisms is unclear. We examined whether the beneficial effects of the imidazoline derivative RX 871024 (RX) on beta-cell function might be related to perturbations of islet NO production. METHODS: Experiments were performed with isolated islets or intact mice challenged with glucose or carbachol with or without RX treatment. Insulin was determined with radioimmunoassay, NO generation with high-performance liquid chromatography and expression of inducible NO synthase (iNOS) with confocal microscopy. RESULTS: RX treatment, in doses lacking effects on basal insulin, greatly amplified insulin release stimulated by the NO-generating secretagogues glucose and carbachol both in vitro and in vivo. RX also improved the glucose tolerance curve. Islets incubated at high glucose levels (20 mmol L(-1)) displayed increased NO production derived from both neuronal constitutive NO synthase (ncNOS) and iNOS. RX abrogated this glucose-induced NO production concomitant with amplification of insulin release. Confocal microscopy revealed abundant iNOS expression in beta cells after incubation of islets at high but not low glucose levels. This was abolished after RX treatment. Similarly, islets cultured for 24 h at high glucose levels showed intense iNOS expression in beta cells. This was abrogated with RX and followed by an amplified glucose-induced insulin release. CONCLUSION: RX effectively counteracts the negative impact of beta-cell NO generation on insulin release stimulated by glucose and carbachol suggesting imidazoline compounds by virtue of NOS inhibitory properties being of potential therapeutic value for treatment of beta-cell dysfunction in hyperglycaemia and type 2 diabetes.


Assuntos
Imidazóis/farmacologia , Indóis/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico Sintase Tipo II/biossíntese , Óxido Nítrico/biossíntese , Animais , Carbacol/farmacologia , Células Cultivadas , Cromatografia Líquida de Alta Pressão/métodos , Relação Dose-Resposta a Droga , Feminino , Glucose/farmacologia , Imidazóis/uso terapêutico , Indóis/uso terapêutico , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Microscopia Confocal
4.
Cell Mol Life Sci ; 65(14): 2256-65, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18560759

RESUMO

Chronic exposure of pancreatic islets to elevated levels of palmitate leads to beta-cell dysfunction. We examined possible involvement of mitogenactivated protein kinases (MAPKs) and caspase-3 in palmitate-induced beta-cell dysfunction and tested the influence of the anti-diabetic drug rosiglitazone (ROZ). Palmitate amplified glucose-stimulated augmentation of intracellular free calcium ([Ca2+]i) and insulin secretion in incubated islets. ROZ suppressed this amplification, whereas it modestly augmented glucose-induced increase in these events. ROZ suppressed short-term palmitate-induced phosphorylation of pro-apoptotic MAPKs, i.e., SAPK/JNK and p38. Long-term islet culturing with palmitate induced inducible nitric oxide synthase (iNOS) and activated SAPK/JNK-p38. ROZ counteracted these effects. Both palmitate and cytokines activated caspase-3 in MIN6c4-cells and isolated islets. ROZ suppressed palmitate- but not cytokine-induced caspase-3 activation. Finally, after palmitate culturing, ROZ reversed the inhibitory effect on glucose-stimulated insulin release. We suggest that ROZ counteracts palmitateinduced deleterious effects on beta-cell function via suppression of iNOS, pro-apoptotic MAPKs and caspase-3 activities, as evidenced by restoration of glucose-stimulated insulin release.


Assuntos
Inibidores de Caspase , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/enzimologia , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Ácido Palmítico/antagonistas & inibidores , Ácido Palmítico/toxicidade , Tiazolidinedionas/farmacologia , Animais , Cálcio/metabolismo , Caspase 3 , Linhagem Celular , Citocinas/farmacologia , Feminino , Hipoglicemiantes/farmacologia , Técnicas In Vitro , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Modelos Biológicos , Rosiglitazona
5.
Horm Metab Res ; 35(1): 48-54, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12669271

RESUMO

Long-term total parenteral nutrition (TPN) is associated with elevated plasma lipids and a marked decrease of glucose-stimulated insulin release. Since nitric oxide (NO) has been shown to modulate negatively the insulin response to glucose, we investigated the influence of TPN-treatment on isoforms of islet NO-synthase (NOS) activities in relation to the effect of glucagon-like peptide-1 (GLP-1), a known activator of glucose-stimulated insulin release. Isolated islets from TPN rats incubated at basal glucose (1 mmol/l) showed a modestly increased insulin secretion accompanied by an enhanced accumulation of islet cAMP and cGMP. In contrast, TPN islets incubated at high glucose (16.7 mmol/l) displayed an impaired insulin secretion and a strong suppression of islet cAMP content. Moreover, islet inducible NOS (iNOS) as well as islet cGMP content were greatly increased in these TPN islets. A dose-response study of GLP-1 with glucose-stimulated islets showed that GLP-1 could overcome and completely restore the impaired insulin release in TPN islets, bringing about a marked increase in islet cAMP accumulation concomitant with heavy suppression of both glucose-stimulated increase in islet cGMP content and the activities of constitutive NOS (cNOS) and iNOS. These effects of GLP-1 were mimicked by dibutyryl-cAMP. The present results show that the impaired insulin response of glucose-stimulated insulin release seen after TPN treatment is normalized by GLP-1. This beneficial effect of GLP-1 is most probably exerted by a cAMP-induced suppression of both iNOS and cNOS activities in these TPN islets.


Assuntos
Glucagon/farmacologia , Ilhotas Pancreáticas/enzimologia , Óxido Nítrico Sintase/metabolismo , Nutrição Parenteral Total , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Animais , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Peptídeo 1 Semelhante ao Glucagon , Glucose/farmacologia , Insulina/metabolismo , Masculino , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Ratos , Ratos Sprague-Dawley
6.
Eur Surg Res ; 33(2): 86-91, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11399874

RESUMO

The purpose of this study was to investigate the influence of selective denervation of the rat pancreas on hormone secretion and on peripheral insulin sensitivity. Thirteen rats, 7 denervated and 6 sham operated, received an intravenous glucose challenge for 30 min. The basal plasma levels of insulin, glucagon and glucose did not differ between the two groups. An augmented insulin response to glucose was detected in the denervated group, whereas the glucagon response was unaffected. Glucose tolerance was marginally improved. Twenty-four rats, 12 denervated and 12 sham operated, received a constant infusion of glucose, insulin, epinephrine and propranolol in order to inhibit the endogenous insulin release and thus evaluate insulin sensitivity. No significant change in insulin sensitivity could be detected during our experimental conditions. We conclude that selective denervation brings about an increased insulin response to glucose, probably by interrupting a catecholaminergic negative tone on the beta-cell. The sympathectomized animals did not disclose any apparent changes in peripheral insulin sensitivity.


Assuntos
Denervação , Ilhotas Pancreáticas/metabolismo , Pâncreas/inervação , Animais , Glicemia/análise , Glucagon/sangue , Glucose/farmacologia , Insulina/sangue , Resistência à Insulina , Masculino , Ratos , Ratos Sprague-Dawley , Valores de Referência
7.
Am J Physiol Endocrinol Metab ; 281(1): E171-9, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11404235

RESUMO

We examined the relation between nutrient-stimulated insulin secretion and the islet lysosome acid glucan-1,4-alpha-glucosidase system in rats undergoing total parenteral nutrition (TPN). During TPN treatment, serum glucose was normal, but free fatty acids, triglycerides, and cholesterol were elevated. Islets from TPN-infused rats showed increased basal insulin release, a normal insulin response to cholinergic stimulation but a greatly impaired response when stimulated by glucose or alpha-ketoisocaproic acid. This impairment of glucose-stimulated insulin release was only slightly ameliorated by the carnitine palmitoyltransferase 1 inhibitor etomoxir. However, in parallel with the impaired insulin response to glucose, islets from TPN-infused animals displayed reduced activities of islet lysosomal enzymes including the acid glucan-1,4-alpha-glucosidase, a putative key enzyme in nutrient-stimulated insulin release. By comparison, the same lysosomal enzymes were increased in liver tissue. Furthermore, in intact control islets, the pseudotetrasaccharide acarbose, a selective inhibitor of acid alpha-glucosidehydrolases, dose dependently suppressed islet acid glucan-1,4-alpha-glucosidase and acid alpha-glucosidase activities in parallel with an inhibitory action on glucose-stimulated insulin secretion. By contrast, when incubated with intact TPN islets, acarbose had no effect on either enzyme activity or glucose-induced insulin release. Moreover, when acarbose was added directly to TPN islet homogenates, the dose-response effect on the catalytic activity of the acid alpha-glucosidehydrolases was shifted to the right compared with control homogenates. We suggest that a general dysfunction of the islet lysosomal/vacuolar system and reduced catalytic activities of acid glucan-1,4-alpha-glucosidase and acid alpha-glucosidase may be important defects behind the impairment of the transduction mechanisms for nutrient-stimulated insulin release in islets from TPN-infused rats.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Lisossomos/fisiologia , Nutrição Parenteral Total/efeitos adversos , Acarbose/farmacologia , Fosfatase Ácida/metabolismo , Animais , Inibidores Enzimáticos/farmacologia , Ácidos Graxos não Esterificados/sangue , Glucana 1,4-alfa-Glucosidase/antagonistas & inibidores , Glucosidases/metabolismo , Técnicas In Vitro , Insulina/sangue , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/enzimologia , Lisossomos/enzimologia , Masculino , Ratos , Ratos Sprague-Dawley , Triglicerídeos/sangue
8.
Am J Physiol Cell Physiol ; 280(5): C1242-54, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11287338

RESUMO

We investigated, by a combined in vivo and in vitro approach, the temporal changes of islet nitric oxide synthase (NOS)-derived nitric oxide (NO) and heme oxygenase (HO)-derived carbon monoxide (CO) production in relation to insulin and glucagon secretion during acute endotoxemia induced by lipopolysaccharide (LPS) in mice. Basal plasma glucagon, islet cAMP and cGMP content after in vitro incubation, the insulin response to glucose in vivo and in vitro, and the insulin and glucagon responses to the adenylate cyclase activator forskolin were greatly increased after LPS. Immunoblots demonstrated expression of inducible NOS (iNOS), inducible HO (HO-1), and an increased expression of constitutive HO (HO-2) in islet tissue. Immunocytochemistry revealed a marked expression of iNOS in many beta-cells, but only in single alpha-cells after LPS. Moreover, biochemical analysis showed a time dependent and markedly increased production of NO and CO in these islets. Addition of a NOS inhibitor to such islets evoked a marked potentiation of glucose-stimulated insulin release. Finally, after incubation in vitro, a marked suppression of NO production by both exogenous CO and glucagon was observed in control islets. This effect occurred independently of a concomitant inhibition of guanylyl cyclase. We suggest that the impairing effect of increased production of islet NO on insulin secretion during acute endotoxemia is antagonized by increased activities of the islet cAMP and HO-CO systems, constituting important compensatory mechanisms against the noxious and diabetogenic actions of NO in endocrine pancreas.


Assuntos
Endotoxemia/fisiopatologia , Heme Oxigenase (Desciclizante)/metabolismo , Ilhotas Pancreáticas/fisiopatologia , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Animais , Glicemia/metabolismo , Monóxido de Carbono/metabolismo , Colforsina/farmacologia , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Endotoxemia/enzimologia , Feminino , Glucagon/sangue , Glucagon/farmacologia , Glucose/farmacologia , Técnicas In Vitro , Insulina/sangue , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/enzimologia , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos , NG-Nitroarginina Metil Éster/farmacologia , Salmonella typhimurium , Fatores de Tempo
9.
Endocrine ; 16(2): 97-104, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11887940

RESUMO

The expression and activities of constitutive nitric oxide synthase (cNOS) and inducible nitric oxide synthase (iNOS) in relation to insulin and glucagon secretory mechanisms were investigated in islets isolated from rats subjected to total parenteral nutrition (TPN) for 10 d. TPN is known to result in significantly increased levels of plasma lipids during the infusion time. In comparison with islets from freely fed control rats, islets taken from TPN rats at d 10 displayed a marked decrease in glucose-stimulated insulin release (4.65 +/- 0.45 ng/[islet x h] vs 10.25 +/- 0.65 for controls) (p < 0.001) accompanied by a strong iNOS activity (18.3 +/- 1.1 pmol of NO/[min x mg of protein]) and a modestly reduced cNOS activity (11.3 +/- 3.2 pmol of NO/[min x mg of protein] vs 17.7 +/- 1.7 for controls) (p < 0.01). Similarly, Western blots showed the expression of iNOS protein as well as a significant reduction in cNOS protein in islets from TPN-treated rats. The enhanced NO production, which is known to inhibit glucose-stimulated insulin release, was manifested as a strong increase in the cyclic guanosine 5'-monophosphate content in the islets of TPN-treated rats (1586 +/- 40 amol/islet vs 695 +/- 64 [p < 0.001] for controls). Moreover, the content of cyclic adenosine monophosphate (cAMP) was greatly increased in the TPN islets (80.4 +/- 2.1 fmol/islet vs 42.6 +/- 2.6 [p < 0.001] for controls). The decrease in glucose-stimulated insulin release was associated with an increase in the activity of the secretory pathway regulated by the cAMP system in the islets of TPN-treated rats, since the release of insulin stimulated by the phosphodiesterase inhibitor isobutylmethylxanthine was greatly increased both in vivo after iv injection and after in vitro incubation of isolated islets. By contrast, the release of glucagon was clearly reduced in islets taken from TPN-treated rats (33.5 +/- 1.5 pg/[islet x h] vs 45.5 +/- 2.2 for controls) (p < 0.01) when islets were incubated at low glucose (1.0 mmol/L). The data show that long-term TPN treatment in rats brings about impairment of glucose-stimulated insulin release, that might be explained by iNOS expression and a marked iNOS-derived NO production in the beta-cells. The release of glucagon, on the other hand, is probably decreased by a direct "nutrient effect" of the enhanced plasma lipids. The results also suggest that the islets of TPN-treated rats have developed compensatory insulin secretory mechanisms by increasing the activity of their beta-cell cAMP system.


Assuntos
Hormônios/metabolismo , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico Sintase/metabolismo , Nutrição Parenteral Total , 1-Metil-3-Isobutilxantina/administração & dosagem , 1-Metil-3-Isobutilxantina/farmacologia , Animais , AMP Cíclico/metabolismo , GMP Cíclico/metabolismo , Glucagon/metabolismo , Glucose/farmacologia , Técnicas In Vitro , Injeções Intravenosas , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Sprague-Dawley , Valores de Referência
10.
Am J Physiol Endocrinol Metab ; 279(1): E95-E107, 2000 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10893328

RESUMO

Islet production of nitric oxide (NO) and CO in relation to islet hormone secretion was investigated in mice given the NO synthase (NOS) inhibitor N(G)-nitro-L-arginine methyl ester (L-NAME) in their drinking water. In these mice, the total islet NO production was paradoxically increased, reflecting induction of inducible NOS (iNOS) in background of reduced activity and immunoreactivity of constitutive NOS (cNOS). Unexpectedly, normal mice fasted for 24 h also displayed iNOS activity, which was further increased in L-NAME-drinking mice. Glucose-stimulated insulin secretion in vitro and in vivo was increased in fasted but unaffected in fed mice after L-NAME drinking. Glucagon secretion was increased in vitro. Control islets incubated with different NOS inhibitors at 20 mM glucose displayed increased insulin release and decreased cNOS activity. These NOS inhibitors potentiated glucose-stimulated insulin release also from islets of L-NAME-drinking mice. In contrast, glucagon release was suppressed. In islets from L-NAME-drinking mice, cyclic nucleotides were upregulated, and forskolin-stimulated hormone release, CO production, and heme oxygenase (HO)-2 expression increased. In conclusion, chronic NOS blockade evoked iNOS-derived NO production in pancreatic islets and elicited compensatory mechanisms against the inhibitory action of NO on glucose-stimulated insulin release by inducing upregulation of the islet cAMP and HO-CO systems.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico/biossíntese , Animais , Glicemia/análise , Cálcio/fisiologia , Calmodulina/fisiologia , Monóxido de Carbono/metabolismo , Cerebelo/metabolismo , Jejum/sangue , Feminino , Glucagon/sangue , Glucose/farmacologia , Indazóis/farmacologia , Insulina/sangue , Secreção de Insulina , Camundongos , Camundongos Endogâmicos , Nucleotídeos Cíclicos/metabolismo , Valores de Referência , Fatores de Tempo
11.
J Endocrinol ; 165(2): 293-300, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10810293

RESUMO

The mechanism of nutrient-evoked insulin release is clearly complex. One part of that mechanism is postulated to be the activation of the glycogenolytic enzyme acid glucan-1,4-alpha-glucosidase. As nitric oxide (NO) has been found to be a potent inhibitor of glucose-stimulated insulin secretion, we have now investigated a possible influence of exogenous NO and inhibition of endogenous NO production on islet acid glucan-1,4-alpha-glucosidase activity in relation to insulin release stimulated by glucose and l-arginine. In isolated islets, NO derived from the intracellular NO donor hydroxylamine inhibited the activation of acid glucan-1, 4-alpha-glucosidase and its isoform acid alpha-glucosidase in parallel with inhibition of glucose-stimulated insulin release. In comparison, other lysosomal enzymes were largely unaffected. Similarly, the spontaneous NO donor sodium nitroprusside, as well as NO gas, when added to islet homogenates, suppressed the activities of these acid alpha-glucosidehydrolases and, to a lesser extent, the activities of other lysosomal enzymes. Finally, in the presence of the NO synthase inhibitor N(G)-nitro-l-arginine methyl ester, insulin release from isolated islets stimulated by glucose or l-arginine was markedly potentiated in parallel with an accompanying increase in the activities of acid glucan-1,4-alpha-glucosidase and acid alpha-glucosidase. Other lysosomal enzymes and neutral alpha-glucosidase were not influenced. We propose that an important inhibitory effect of NO on the insulin secretory processes stimulated by glucose and l-arginine is exerted via inactivation of islet acid glucan-1,4-alpha-glucosidase, a putative key enzyme in nutrient-stimulated insulin release.


Assuntos
Inibidores Enzimáticos/farmacologia , Glucana 1,4-alfa-Glucosidase/antagonistas & inibidores , Ilhotas Pancreáticas/enzimologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico/farmacologia , Animais , Técnicas de Cultura , Feminino , Glucana 1,4-alfa-Glucosidase/metabolismo , Inibidores de Glicosídeo Hidrolases , Hidroxilamina/farmacologia , Camundongos , Camundongos Endogâmicos , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Nitroprussiato/farmacologia , alfa-Glucosidases/metabolismo
12.
Pancreas ; 20(2): 170-6, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10707933

RESUMO

Pancreaticobiliary diversion (PBD) and biliodigestive shunt (BDS) cause long-standing hypercholecystokininemia followed by pancreatic hyperplasia. These changes have been suggested to be due to the lack of intraluminal trypsin and bile, respectively, in the upper small intestine. The aim of these experiments was to study the effect of restoration of intraluminal trypsin and bile on plasma levels of cholecystokinin (CCK) and the changes found in exocrine and endocrine pancreas after PBD and BDS. Male Sprague-Dawley rats were used. PBD was done in 16 rats, eight of which had trypsin dissolved in 50 mM sodium bicarbonate (SB), and eight had SB only by gastric intubation twice daily. BDS was done in another 16 rats, eight of which had bile dissolved in SB, and eight had SB in a similar manner. Sham-operated rats had SB and served as controls. After 4 weeks, the rats were killed, and the concentrations of circulating CCK, gastrin, glucose, glucagon, and insulin were determined. The pancreas was removed, weighed, and analyzed for contents of water, protein, and DNA. In another study, PBD-operated rats got trypsin in varying dosages or trypsin and taurocholate in combination for 2 weeks before death. The concentrations of plasma CCK and glucagon were elevated after both PBD and BDS. PBD decreased the concentration of gastrin in plasma. PBD caused an increase of pancreatic weight and the contents of protein and DNA. Trypsin substitution to PBD-operated rats did not affect plasma CCK or glucagon levels, but the PBD-induced increases in weight and DNA content were counteracted by trypsin. Higher dosages of trypsin did not further influence the effects seen after PBD. Pancreatic weight and DNA content were increased after BDS. Bile administration completely abolished the increase in plasma CCK and glucagon, as well as the gain in pancreatic weight, and reduced the increase in pancreatic DNA. Substitution with bile to BDS-operated rats abolished the increase in the plasma levels of CCK and glucagon, as well as the trophic effects on the pancreas. Trypsin substitution to PBD-operated rats partly reversed the trophic effects on the pancreas but not the hormonal changes in plasma. Thus the trophic effects on the pancreas exerted by BDS seem to be dependent on the lack of bile in the upper small intestine, whereas the effects of PBD only partly are a consequence of the absence of intraluminal trypsin.


Assuntos
Bile/fisiologia , Desvio Biliopancreático , Procedimentos Cirúrgicos do Sistema Digestório , Ilhotas Pancreáticas/fisiologia , Pâncreas/fisiologia , Tripsina/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Colecistocinina/sangue , Relação Dose-Resposta a Droga , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Pâncreas/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Tripsina/farmacologia
13.
Diabetologia ; 42(8): 978-86, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10491758

RESUMO

AIMS/HYPOTHESIS: To map the cellular location of inducible and constitutive nitric oxide synthase and haem oxygenase in rat islets to clarify the morphological background to putative nitric oxide and carbon monoxide pathways. METHODS: Immunocytochemistry and confocal microscopy. RESULTS: After treatment with endotoxin, immunoreactivity for inducible nitric oxide synthase was expressed in a large number of islet cells, most of which were insulin-immunoreactive beta cells and in single glucagon-immunoreactive and pancreatic polypeptide-immunoreactive cells. Somatostatin-immunoreactive cells lacked immunoreactivity for inducible nitric oxide synthase. In untreated rats, immunoreactivity for constitutive nitric oxide synthase occurred in the majority of insulin-immunoreactive and glucagon-immunoreactive cells, in most pancreatic polypeptide-immunoreactive and somatostatin-immunoreactive cells and in islet nerves. Similarly, immunoreactivity for constitutive haem oxygenase was detected in all four types of islet cells. Endotoxin treatment did not change the pattern of immunoreactivity for constitutive and inducible haem oxygenase. After treatment with alloxan, insulin-immunoreactivity was observed only in single islet cells, being almost devoid of immunoreactivity for constitutive nitric oxide synthase and haem oxygenase. CONCLUSION/INTERPRETATION: In vivo endotoxin-induced expression of inducible nitric oxide synthase in insulin-producing and in scattered glucagon-producing and pancreatic polypeptide-producing cells strengthens previous suggestions of a pathophysiological role for inducible nitric oxide synthase in the development of insulin-dependent diabetes mellitus. The presence of constitutive nitric oxide synthase and haem oxygenase in all four types of islet cells, together with recent functional data of ours support roles for nitric oxide and carbon monoxide as intracellular, paracrine or neurocrine modulators of islet hormone secretion.


Assuntos
Monóxido de Carbono/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico/metabolismo , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Endotoxinas/toxicidade , Feminino , Heme Oxigenase (Desciclizante)/metabolismo , Imuno-Histoquímica/métodos , Ilhotas Pancreáticas/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Microscopia Confocal/métodos , Óxido Nítrico Sintase Tipo I , Óxido Nítrico Sintase Tipo II , Ratos , Ratos Sprague-Dawley , Salmonella typhimurium
14.
J Endocrinol ; 163(1): 39-48, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10495405

RESUMO

We have studied, by a combined in vitro and in vivo approach, the relation between the inhibitory action of N(G)-nitro-l-arginine methyl ester (L-NAME), a selective inhibitor of nitric oxide synthase (NOS), on the activity of islet constitutive NOS (cNOS) and glucose regulation of islet hormone release in mice. The cNOS activity in islets incubated in vitro at 20 mM glucose was not appreciably affected by 0.05 or 0.5 mM L-NAME, but was greatly suppressed (-60%) by 5 mM L-NAME. Similarly, glucose-stimulated insulin release was unaffected by the lower concentrations of L-NAME but greatly enhanced in the presence of 5 mM of the NOS inhibitor. In incubated islets inhibition of cNOS activity resulted in a modestly enhanced insulin release in the absence of glucose, did not display any effect at physiological or subphysiological glucose concentrations, but resulted in a markedly potentiated insulin release at hyperglycaemic glucose concentrations. In the absence of glucose, glucagon secretion was suppressed by L-NAME. The dynamics of glucose-induced insulin release and (45)Ca(2+) efflux from perifused islets revealed that L-NAME caused an immediate potentiation of insulin release, and a slight increase in (45)Ca(2+) efflux. In islets depolarized with 30 mM K(+) in the presence of the K(+)(ATP) channel opener, diazoxide, L-NAME still greatly potentiated glucose-induced insulin release. Finally, an i.v. injection of glucose to mice pretreated with L-NAME was followed by a markedly potentiated insulin response, and an improved glucose tolerance. In accordance, islets isolated directly ex vivo after L-NAME injection displayed a markedly reduced cNOS activity. In conclusion, we have shown here, for the first time, that biochemically verified suppression of islet cNOS activity, induced by the NOS inhibitor L-NAME, is accompanied by a marked potentiation of glucose-stimulated insulin release both in vitro and in vivo. The major action of NO to inhibit glucose-induced insulin release is probably not primarily linked to changes in Ca(2+) fluxes and is exerted mainly independently of membrane depolarization events.


Assuntos
Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase/metabolismo , Análise de Variância , Animais , Cálcio/metabolismo , Técnicas de Cultura , Diazóxido/farmacologia , Inibidores Enzimáticos/farmacologia , Feminino , Glucagon/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Endogâmicos , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase Tipo II , Óxido Nítrico Sintase Tipo III , Canais de Potássio/efeitos dos fármacos
15.
Endocrinology ; 140(7): 3045-53, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10385396

RESUMO

Accumulated evidence links an important signal involved in glucose-stimulated insulin release to the activation of the islet lysosomal glycogenolytic enzyme acid glucan-1,4-alpha-glucosidase. We have analyzed the function of the lysosomal system/lysosomal enzyme activities in pancreatic islets of young (6-8 weeks), spontaneously diabetic, GK (Goto-Kakizaki) rats and Wistar control rats in relation to glucose-induced insulin release. The insulin secretory response to glucose was markedly impaired in the GK rat, but was restored by the adenylate cyclase activator forskolin. Islet activities of classical lysosomal enzymes, e.g.. acid phosphatase, N-acetyl-beta-D-glucosaminidase, beta-glucuronidase, and cathepsin D, were reduced by 20-35% in the GK rat compared with those in Wistar controls. In contrast, the activities of the lysosomal alpha-glucosidehydrolases, i.e.. acid glucan-1,4-alpha-glucosidase and acid alpha-glucosidase, were increased by 40-50%. Neutral alpha-glucosidase (endoplasmic reticulum) was unaffected. Comparative analysis of liver tissue showed that lysosomal enzyme activities were of the same magnitude in GK and Wistar rats. Notably, in Wistar rats, the activities of acid glucan-1,4-alpha-glucosidase and acid alpha-glucosidase were approximately 15-fold higher in islets than in liver. Other lysosomal enzymes did not display such a difference. Normalization of glycemia in GK rats by phlorizin administered for 9 days did not influence either the lysosomal alpha-glucosidehydrolase activities or other lysosomal enzyme activities in GK islets. Finally, the pseudotetrasaccharide acarbose, which accumulates in the lysosomal system, inhibited acid glucan-1,4-alpha-glucosidase activity in parallel with its inhibitory action on glucose-induced insulin release in intact Wistar islets, whereas no effect was recorded for either parameter in intact GK islets. In contrast, acarbose inhibited the enzyme activity equally in islet homogenates from both GK and Wistar rats, showing that the catalytic activity of the enzyme itself in disrupted cells was unaffected. We propose that dysfunction of the islet lysosomal/vacuolar system is an important defect impairing the transduction mechanisms for glucose-induced insulin release in the GK rat.


Assuntos
Diabetes Mellitus/metabolismo , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Lisossomos/metabolismo , Acarbose , Animais , Colforsina/farmacologia , Diabetes Mellitus/genética , Inibidores Enzimáticos/farmacologia , Glucana 1,4-alfa-Glucosidase/antagonistas & inibidores , Técnicas In Vitro , Secreção de Insulina , Fígado/enzimologia , Lisossomos/enzimologia , Florizina/farmacologia , Ratos , Ratos Endogâmicos/genética , Ratos Wistar , Trissacarídeos/farmacologia
16.
Pancreas ; 18(3): 266-73, 1999 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10206485

RESUMO

Obese mice (Umeå ob/ob) and their lean litter-mates were investigated from 7 to 52 weeks of age with respect to the plasma concentration of islet amyloid polypeptide (IAPP) and insulin. Plasma levels of IAPP were highly elevated in the ob/ob mice and remained unchanged until age 33 weeks, after which a sudden significant increase occurred at age 40 weeks. The plasma concentration of insulin gradually increased from start to end and reached extremely high levels. In the lean mice, there were no age-related differences in plasma levels of IAPP and insulin, being of the same magnitude as in normal NMRI mice. The plasma IAPP/insulin molar ratio was similar in lean and obese mice until age 14 weeks. At 21 weeks, the ratio in the ob/ob mice had decreased dramatically and remained markedly (sixfold) lower than in the lean mice until the end of the study. The IAPP concentration in the pancreata of 21-week-old ob/ob mice was 25-fold higher than that in the lean mice. Immunohistochemically, a majority of the ob/ob mice displayed enlarged and more numerous pancreatic islets, compared with the lean mice, and the IAPP- and insulin-labeling intensity was equal for all animals. At the electron-microscopic level, there was an increase in the number of IAPP- and insulin-immunoreactive gold particles per whole granule area as well as per core granule area. We conclude that the dramatically increased IAPP levels in severely hyperinsulinemic ob/ob mice may be of importance for the development of insulin resistance. Further, the disproportionate secretion of IAPP and insulin in the adult obese mouse might indicate a disturbed negative feedback effect of IAPP on insulin secretory mechanisms, resulting in very high plasma insulin levels.


Assuntos
Amiloide/sangue , Insulina/sangue , Obesidade/sangue , Envelhecimento , Animais , Feminino , Imuno-Histoquímica , Polipeptídeo Amiloide das Ilhotas Pancreáticas , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Obesos , Microscopia Imunoeletrônica , Obesidade/patologia
17.
J Physiol ; 515 ( Pt 2): 463-73, 1999 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-10050013

RESUMO

1. We have investigated, with a combined in vitro and in vivo approach, the influence on insulin and glucagon release stimulated by the cholinergic, muscarinic agonist carbachol of different NO modulators, i.e. the nitric oxide synthase (NOS) inhibitors NG-nitro-L-arginine methyl ester (L-NAME), NG-monomethyl-L-arginine (L-NMMA) and 7-nitroindazole as well as the intracellular NO donor hydroxylamine. 2. At basal glucose (7 mM) carbachol dose-dependently stimulated insulin release from isolated islets with a half-maximal response at approximately 1 microM of the agonist. In the presence of 5 mM L-NAME (a concentration that did not influence basal insulin release) the insulin response was markedly increased along the whole dose-response curve and the threshold for carbachol stimulation was significantly lowered. 3. Carbachol-stimulated islets displayed an increased insulin release and a suppressed glucagon release in the presence of L-NAME, L-NMMA or 7-nitroindazole. Significant suppression of glucagon release (except for L-NAME) was achieved at lower concentrations (approximately 0.1-0.5 mM) of the NOS inhibitors than the potentiation of insulin release (1.0-5.0 mM). The intracellular NO donor hydroxylamine dose-dependently inhibited carbachol-induced insulin release but stimulated glucagon release only at a low concentration (3 microM). 4. In islets depolarized with 30 mM K+ in the presence of the KATP channel opener diazoxide, NOS inhibition by 5 mM L-NAME still markedly potentiated carbachol-induced insulin release (although less so than in normal islets) and suppressed glucagon release. 5. In vivo pretreatment of mice with L-NAME was followed by a markedly increased insulin release and a reduced glucagon release in response to an i.v. injection of carbachol. 6. The data suggest that NO is a negative modulator of insulin release but a positive modulator of glucagon release induced by cholinergic muscarinic stimulation. These effects were also evident in K+ depolarized islets and thus NO might exert a major influence on islet hormone secretion independently of membrane depolarization events.


Assuntos
Carbacol/farmacologia , Agonistas Colinérgicos/farmacologia , Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico/fisiologia , Animais , Relação Dose-Resposta a Droga , Eletrofisiologia , Inibidores Enzimáticos/farmacologia , Feminino , Hidroxilamina/farmacologia , Indazóis/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Endogâmicos , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Potássio/farmacologia , ômega-N-Metilarginina/farmacologia
18.
Diabetes ; 48(1): 66-76, 1999 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-9892224

RESUMO

Carbon monoxide (CO) has been suggested as a novel messenger molecule in the brain. We now report on the cellular localization and hormone secretory function of a CO-producing constitutive heme oxygenase (HO-2) in mouse islets. Islet homogenates produced large amounts of CO which were suppressed dose-dependently by the HO inhibitor zincprotoporphyrin-IX (ZnPP-IX). We also show, for the first time, that glucose markedly stimulates the HO activity (CO production) in intact islets. A further potentiation was induced by the HO substrate hemin. Western blot showed that islet tissue expressed HO-2, and confocal microscopy revealed that HO-2 resided in insulin, glucagon, somatostatin, and pancreatic polypeptide cells. ZnPP-IX dose-dependently inhibited, whereas hemin enhanced, both insulin and glucagon secretion from glucose-stimulated islets. Stimulation or inhibition of CO production was accompanied by corresponding changes in islet cGMP levels. Exogenously applied CO stimulated insulin and glucagon release from isolated islets, whereas exogenous nitric oxide (NO) inhibited insulin and stimulated glucagon release. Islets stimulated by glucose or L-arginine displayed a marked increase in their NO-synthase (NOS) activity. Such an increase was suppressed by hemin, conceivably because NOS activity was inhibited by hemin-derived CO. Consequently, hemin enhanced L-arginine-induced insulin secretion. Insulin release stimulated by either hemin-derived CO or exogenous CO was strongly inhibited by the guanylate cyclase inhibitor ODQ, but it was unaffected by ZnPP-IX. Glucagon release induced by CO (but not by hemin) was inhibited by ODQ and partly inhibited by ZnPP-IX. We propose that the islets of Langerhans are equipped with a heme oxygenase-carbon monoxide pathway, which constitutes a novel regulatory system of physiological importance for the stimulation of insulin and glucagon release. This pathway is stimulated by glucose, is at least partly dependent on the cGMP system, and displays interaction with islet NOS activity.


Assuntos
Monóxido de Carbono/metabolismo , Heme Oxigenase (Desciclizante)/metabolismo , Hormônios/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Western Blotting , Monóxido de Carbono/farmacologia , Monóxido de Carbono/fisiologia , GMP Cíclico/metabolismo , Interações Medicamentosas , Inibidores Enzimáticos/farmacologia , Feminino , Glucose/farmacologia , Hemina/farmacologia , Imuno-Histoquímica , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Microscopia Confocal , Óxido Nítrico/fisiologia , Oxidiazóis/farmacologia , Protoporfirinas/farmacologia , Quinoxalinas/farmacologia
19.
J Physiol ; 514 ( Pt 2): 579-91, 1999 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9852337

RESUMO

1. Mice were subjected to gastrectomy (GX) or food deprivation (24 h). The release of insulin and glucagon in response to different secretagogues was monitored in vivo and in isolated islets 3-4 weeks after surgery. 2. GX animals responded to glucose with an impaired glucose tolerance and a poor increase in plasma insulin. Islets from GX or food-deprived mice displayed impaired insulin release to high glucose and enhanced glucagon release at low glucose. 3. After GX the insulinogenic index, Delta insulin (microU ml-1)/Delta glucose (mg ml-1), was suppressed by 65% after oral glucose and by 59% after i.v. glucose. The integrated insulin response after oral glucose was reduced by 90% in GX mice. After i.v. glucose the reduction was 67%. 4. Carbachol-induced insulin release in vivo was reduced after food deprivation and exaggerated after GX. Carbachol-stimulated glucagon secretion was suppressed after GX and after food deprivation. A similar pattern was found in vitro. 5. Cyclic AMP activation (by the phosphodiesterase inhibitor isobutylmethylxanthine or the adenylate cyclase stimulator forskolin) induced a greater insulin response in GX or food-deprived mice than in sham-operated, fed mice. A similar pattern was found in vitro. The glucagon response was enhanced in vitro but not in vivo. 6. Crude extracts of rat oxyntic mucosa enhanced basal as well as glucose-induced insulin release from isolated islets, whereas glucagon release was markedly inhibited. The effects were dose dependent, the inhibition of glucagon release being achieved at lower concentrations than the potentiation of glucose-induced insulin release. The active principle was inactivated by incubation with trypsin or leucine aminopeptidase. 7. The data suggest that a circulating agent, probably a peptide, from gastric oxyntic mucosa stimulates glucose-induced insulin secretion. It also suppresses glucagon secretion. The GX-evoked impairment of the insulin (and glucagon) response to glucose is partly compensated for by an enhanced insulin response to cholinergic and/or cyclic AMP activation.


Assuntos
AMP Cíclico/metabolismo , Gastrectomia , Glucagon/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Células Parietais Gástricas/fisiologia , Estômago/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Animais , Glicemia/metabolismo , Carbacol/farmacologia , Colforsina/farmacologia , Jejum , Feminino , Privação de Alimentos , Glucagon/sangue , Glucose/farmacologia , Insulina/sangue , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos , Ratos , Extratos de Tecidos/farmacologia , Vagotomia
20.
Endocrine ; 11(1): 99-107, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10668648

RESUMO

We have investigated the influence of the intracellular free radical donors hydroxylamine (giving nitric oxide [NO]) and tert-butylhydroperoxide (giving hydroperoxide ["H2O2"]) on glucose- and cyclic adenosine monophosphate (cAMP)-induced transduction signaling in islet hormone release. Both donors dose dependently inhibited glucose-stimulated insulin release and induced modest (hydroxylamine) or profound (tertbutylhydroperoxide) suppression of 45Ca2+-efflux from perifused islets. By contrast, both donors stimulated glucagon release. Similar effects on hormone release were displayed after K+-depolarization. Insulin and glucagon release stimulated by activation of the cAMP system through isobutylmethylxanthine (IBMX) at basal glucose was modestly potentiated by low concentrations of both donors. These effects were still observed, although less pronounced, in K+-depolarized islets. In vitro as well as in vivo, the NO-synthase inhibitor N(G)-nitro-L-arginine methyl ester inhibited IBMX-induced glucagon release, but did not affect insulin release. The results suggest that NO and hydroperoxide inhibit glucose-stimulated insulin release by perturbing Ca2+ fluxes and probably acting through S-nitrosylation (NO) or oxidation (hydroperoxide) of thiol groups critical to the secretory process. These effects are largely independent of depolarization events. By contrast, both NO and hydroperoxide can potentiate cAMP-stimulated hormone release presumably at a distal site in the stimulus-secretion coupling.


Assuntos
Cálcio/metabolismo , Peróxido de Hidrogênio/farmacologia , Ilhotas Pancreáticas/metabolismo , Óxido Nítrico/fisiologia , Oxidantes/farmacologia , 1-Metil-3-Isobutilxantina/farmacologia , Transportadores de Cassetes de Ligação de ATP , Animais , Catalase/antagonistas & inibidores , Catalase/metabolismo , AMP Cíclico/metabolismo , Inibidores Enzimáticos/farmacologia , Radicais Livres/metabolismo , Glucagon/metabolismo , Glucose/metabolismo , Glucose/farmacologia , Glutationa Peroxidase/antagonistas & inibidores , Glutationa Peroxidase/metabolismo , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Canais KATP , Masculino , Camundongos , NG-Nitroarginina Metil Éster/farmacologia , Doadores de Óxido Nítrico/farmacologia , Óxido Nítrico Sintase/antagonistas & inibidores , Canais de Potássio/agonistas , Canais de Potássio Corretores do Fluxo de Internalização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA