Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 127
Filtrar
1.
J Exp Med ; 181(2): 559-68, 1995 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-7530759

RESUMEN

Cytokines, released in and around pancreatic islets during insulitis, have been proposed to participate in beta-cell destruction associated with autoimmune diabetes. In this study we have evaluated the hypothesis that local release of the cytokine interleukin 1 (IL-1) by nonendocrine cells of the islet induce the expression of inducible nitric oxide synthase (iNOS) by beta cells which results in the inhibition of beta cell function. Treatment of rat islets with a combination of tumor necrosis factor (TNF) and lipopolysaccharide (LPS), conditions known to activate macrophages, stimulate the expression of iNOS and the formation of nitrite. Although TNF+LPS induce iNOS expression and inhibit insulin secretion by intact islets, this combination does not induce the expression of iNOS by beta or alpha cells purified by fluorescence activated cell sorting (Facs). In contrast, IL-1 beta induces the expression of iNOS and also inhibits insulin secretion by both intact islets and Facs-purified beta cells, whereas TNF+LPS have no inhibitory effects on insulin secretion by purified beta cells. Evidence suggests that TNF+LPS inhibit insulin secretion from islets by stimulating the release of IL-1 which subsequently induces the expression of iNOS by beta cells. The IL-1 receptor antagonist protein completely prevents TNF+LPS-induced inhibition of insulin secretion and attenuates nitrite formation from islets, and neutralization of IL-1 with antisera specific for IL-1 alpha and IL-1 beta attenuates TNF+LPS-induced nitrite formation by islets. Immunohistochemical localization of iNOS and insulin confirm that TNF+LPS induce the expression of iNOS by islet beta cells, and that a small percentage of noninsulin-containing cells also express iNOS. Local release of IL-1 within islets appears to be required for TNF+LPS-induced inhibition of insulin secretion because TNF+LPS do not stimulate nitrite formation from islets physically separated into individual cells. These findings provide the first evidence that a limited number of nonendocrine cells can release sufficient quantities of IL-1 in islets to induce iNOS expression and inhibit the function of the beta cell, which is selectively destroyed during the development of autoimmune diabetes.


Asunto(s)
Aminoácido Oxidorreductasas/biosíntesis , Interleucina-1/metabolismo , Islotes Pancreáticos/metabolismo , Aminoácido Oxidorreductasas/metabolismo , Animales , Separación Celular , Inducción Enzimática , Citometría de Flujo , Inmunohistoquímica , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/fisiología , Lipopolisacáridos/farmacología , Masculino , Óxido Nítrico Sintasa , Nitritos/metabolismo , Ratas , Ratas Sprague-Dawley
2.
J Cell Biol ; 97(2): 433-7, 1983 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-6350317

RESUMEN

Immunoreactive insulin was shown to exist as a surface molecule in the plasma membrane of dispersed rat pancreatic islet cells. The intact cells were stained by immunofluorescence with a guinea pig antisera specific for insulin. The hormone on the cell surface could not be accounted for by insulin bound to specific receptors or nonspecifically absorbed to cells. Thus, surface insulin was demonstrated to be a specific membrane antigen for islet cells. Furthermore, the proportion of islet cells with insulin on the cell surface was directly correlated with insulin secretion in several different settings. This correspondence was demonstrated by varying the glucose concentration in the medium, by withholding Ca2+, which inhibits secretion, and by adding theophylline, which potentiates secretion. Consequently, these results suggested that insulin as a membrane protein was a marker for cells that actively secreted the hormone and may have been derived in the fusion process of secretory granules with the plasma membrane.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Antígenos de Superficie , Membrana Celular/inmunología , Membrana Celular/metabolismo , Técnicas In Vitro , Insulina/inmunología , Secreción de Insulina , Islotes Pancreáticos/inmunología , Masculino , Ratas , Ratas Endogámicas , Receptor de Insulina/metabolismo
3.
J Clin Invest ; 85(2): 482-90, 1990 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-2405021

RESUMEN

Recent evidence has suggested that pancreatic islets isolated from rats synthesize 1,2-diacyl-sn-glycerol (DAG) de novo from glucose and that this process may constitute the long-sought link between the metabolism of glucose and the induction of insulin secretion. The cell-permeant diacylglycerol 1-oleoyl-2-acetyl-sn-glycerol (200 microM) has been found here to amplify both the first and second phases of insulin secretion from perifused human islets. Measurements of the mass of endogenous DAG in human pancreatic islets by enzymatic and by mass spectrometric methods indicate that levels of 200 microM may be achieved under physiologic conditions. Conversion of [14C]glucose to [14C]DAG has been demonstrated here to occur within 60 s of exposure of rat and human islets to stimulatory concentrations of glucose. This process has been found to be a quantitatively minor contributor to the total islet DAG mass after acute stimulation with glucose, however, and glucose has been found not to induce a rise in total islet DAG content within 20 min of induction of insulin secretion. In contrast to the case with rodent islets, two pharmacologic inhibitors of DAG-induced activation of protein kinase C (staurosporine and sphingosine) have been found not to influence glucose-induced insulin secretion from isolated human islets. These findings indicate that de novo synthesis of DAG from glucose does not participate in acute signal-response coupling in islets.


Asunto(s)
Diglicéridos/biosíntesis , Glucosa/metabolismo , Glicéridos/biosíntesis , Islotes Pancreáticos/metabolismo , Alcaloides/farmacología , Animales , Técnicas de Cultivo , Diglicéridos/análisis , Diglicéridos/farmacología , Glucosa/farmacología , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Perfusión , Proteína Quinasa C/análisis , Ratas , Ratas Endogámicas , Esfingosina/farmacología , Estaurosporina
4.
J Clin Invest ; 90(6): 2384-91, 1992 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-1334975

RESUMEN

Nitric oxide has recently been implicated as the effector molecule that mediates IL-1 beta-induced inhibition of glucose-stimulated insulin secretion and beta-cell specific destruction. The pancreatic islet represents a heterogeneous cell population containing both endocrine cells (beta-[insulin], alpha-]glucagon], gamma[somatostatin], and PP-[polypeptide] secreting cells) and non-endocrine cells (fibroblast, macrophage, endothelial, and dendritic cells). The purpose of this investigation was to determine if the beta-cell, which is selectively destroyed during insulin-dependent diabetes mellitus, is both a source of IL-1 beta-induced nitric oxide production and also a site of action of this free radical. Pretreatment of beta-cells, purified by FACS with IL-1 beta results in a 40% inhibition of glucose-stimulated insulin secretion that is prevented by the nitric oxide synthase inhibitor, NG-monomethyl-L-arginine (NMMA). IL-1 beta induces the formation of nitric oxide by purified beta-cells as evidenced by the accumulation of cGMP, which is blocked by NMMA. IL-1 beta also induces the accumulation of cGMP by the insulinoma cell line Rin-m5F, and both NMMA as well as the protein synthesis inhibitor cycloheximide prevent this cGMP accumulation. Iron-sulfur proteins appear to be intracellular targets of nitric oxide. IL-1 beta induces the formation of an iron-dinitrosyl complex by Rin-m5F cells indicating that nitric oxide mediates the destruction of iron-sulfur clusters of iron containing enzymes. This is further demonstrated by IL-1 beta-induced inhibition of glucose oxidation by purified beta-cells, mitochondrial aconitase activity of dispersed islet cells, and mitochondrial aconitase activity of Rin-m5F cells, all of which are prevented by NMMA. IL-1 beta does not appear to affect FACS-purified alpha-cell metabolic activity or intracellular cGMP levels, suggesting that IL-1 beta does not exert any effect on alpha-cells. These results demonstrate that the islet beta-cell is a source of IL-1 beta-induced nitric oxide production, and that beta-cell mitochondrial iron-sulfur containing enzymes are one site of action of nitric oxide.


Asunto(s)
Interleucina-1/farmacología , Islotes Pancreáticos/metabolismo , Óxido Nítrico/metabolismo , Aconitato Hidratasa/metabolismo , Animales , Arginina/análogos & derivados , Arginina/farmacología , GMP Cíclico/metabolismo , Cicloheximida/farmacología , Espectroscopía de Resonancia por Spin del Electrón , Glucosa/metabolismo , Glucosa/farmacología , Técnicas In Vitro , Insulina/metabolismo , Secreción de Insulina , Masculino , Ratas , Ratas Sprague-Dawley , Tasa de Secreción/efectos de los fármacos , Células Tumorales Cultivadas , omega-N-Metilarginina
5.
J Clin Invest ; 86(3): 856-63, 1990 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-2203826

RESUMEN

Recombinant human IL 1 beta inhibits glucose-induced insulin secretion from isolated pancreatic islets and from purified beta-cells obtained by fluorescence-activated cell sorting (FACS) of dispersed islet cells. Brief (1 h) exposure of isolated islets to IL 1 produces sustained inhibition of insulin secretion for at least 17 h after the IL 1 has been removed from the culture medium. An inhibitory effect of IL 1 on insulin secretion is not observed when islets are coincubated with an inhibitor of DNA transcription (actinomycin D). This finding indicates that the inhibitory effect of IL 1 on insulin secretion requires transcription of one or more genes during the first hour of exposure of islets to IL 1. The inhibitory effect of IL 1 on insulin secretion also requires mRNA translation, because three structurally distinct inhibitors of protein synthesis (cycloheximide, anisomycin, and puromycin) prevent IL 1-induced inhibition of insulin secretion when added to islets after the 1-h exposure to IL 1. Two-dimensional gel electrophoresis of islet proteins metabolically labeled with [35S]methionine demonstrates that IL 1 augments the expression of a 65-kD (pl approximately 6.5) protein by greater than 2.5-fold. These findings indicate that biochemical events occurring within 1 h of exposure of islets to IL 1 lead to an inhibition of insulin secretion that persists for at least 17 h after the removal of IL 1. One of the early biochemical effects of IL 1 on islets is gene transcription (0-1 h), which is followed by mRNA translation (after 1 h). Our results suggest that the inhibitory effect of IL 1 on insulin secretion is mediated by protein(s) whose synthesis is induced by IL 1.


Asunto(s)
Insulina/metabolismo , Interleucina-1/farmacología , Islotes Pancreáticos/metabolismo , Animales , Cicloheximida/farmacología , Dactinomicina/farmacología , Electroforesis en Gel Bidimensional , Expresión Génica/efectos de los fármacos , Glucosa/farmacología , Técnicas In Vitro , Insulina/genética , Secreción de Insulina , Punto Isoeléctrico , Peso Molecular , Biosíntesis de Proteínas/efectos de los fármacos , ARN Mensajero/genética , Ratas , Ratas Endogámicas , Tasa de Secreción/efectos de los fármacos , Factores de Tiempo , Transcripción Genética/efectos de los fármacos
6.
Biochim Biophys Acta ; 834(1): 23-36, 1985 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-3919770

RESUMEN

Isolated pancreatic islets from the rat have been demonstrated by stable isotope dilution-mass spectrometric methods to synthesize the 12-lipoxygenase product 12-hydroxyeicosatetraenoic acid (12-HETE) in amounts of 1.7 to 2.8 ng per 10(3) islets. No detectable amounts of 5-HETE and only trace amounts of 15-HETE could be demonstrated by these methods. Nordihydroguaiaretic acid (NDGA) and BW755C have been demonstrated to inhibit islet 12-HETE synthesis and also to inhibit glucose-induced insulin secretion. Inhibition of insulin secretion and of 12-HETE synthesis exhibited similar dependence on the concentration of these compounds. Eicosa-5,8,11,14-tetrynoic acid (ETYA) also inhibited glucose-induced insulin secretion, as previously reported, at concentrations which inhibit islet 12-HETE synthesis. Exogenous 12-HETE partially reversed the suppression of glucose-induced insulin secretion by lipoxygenase inhibitors, but exogenous 12-hydroperoxyeicosatetraenoic acid (12-HPETE), 15-HPETE, 5-HPETE, 15-HETE, or 5-HETE did not reverse this suppression. These observations argue against the recently suggested hypothesis that islet synthesis of 5-HETE modulates insulin secretion. Suppression of glucose-induced insulin secretion by ETYA, BW755C and NDGA may be due to inhibition of the islet 12-lipoxygenase by these compounds. The possibility that other processes involved in glucose-induced insulin secretion are inhibited by ETYA, BW755C and NDGA cannot yet be excluded.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Leucotrienos , Lipooxigenasa/metabolismo , 4,5-dihidro-1-(3-(trifluorometil)fenil)-1H-pirazol-3-amina , Ácido 5,8,11,14-Eicosatetrainoico/farmacología , Animales , Araquidonato Lipooxigenasas , Ácido Araquidónico , Ácidos Araquidónicos/farmacología , Cromatografía de Gases y Espectrometría de Masas , Ácidos Hidroxieicosatetraenoicos/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Pirazoles/farmacología , Ratas , Ratas Endogámicas
7.
Biochim Biophys Acta ; 729(2): 176-84, 1983 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-6219705

RESUMEN

The properties of active or ATP-dependent calcium transport by islet-cell endoplasmic reticulum and plasma membrane-enriched subcellular fractions were directly compared. These studies indicate that the active calcium transport systems of the two membranes are fundamentally distinct. In contrast to calcium uptake by the endoplasmic reticulum-enriched fraction, calcium uptake by islet-cell plasma membrane-enriched vesicles exhibited a different pH optimum, was not sustained by oxalate, and showed an approximate 30-fold greater affinity for ionized calcium. A similar difference in affinity for calcium was exhibited by the Ca2+-stimulated ATPase activities which are associated with these islet-cell subcellular fractions. Consistent with the effects of calmodulin on calcium transport, calmodulin stimulated Ca2+-ATPase in the plasma membranes, but did not increase calcium-stimulated ATPase activity in the endoplasmic reticulum membranes. The physiological significance of the differences observed in calcium transport by the endoplasmic reticulum and plasma membrane fractions relative to the regulation of insulin secretion by the islets of Langerhans is discussed.


Asunto(s)
Calcio/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Transporte Biológico Activo , ATPasas Transportadoras de Calcio/metabolismo , Calmodulina/farmacología , Membrana Celular/metabolismo , Ácido Egtácico/farmacología , Retículo Endoplásmico/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Oxalatos/farmacología , Ácido Oxálico , Ratas , Ratas Endogámicas
8.
Biochim Biophys Acta ; 794(1): 110-24, 1984 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-6329307

RESUMEN

The metabolism of arachidonic acid by pancreatic islets has been studied with purified populations of large numbers of islets isolated from the rat. Sequential high-performance liquid chromatographic analyses of islet-derived metabolites of 3H-labeled arachidonate in both reversed and normal phases with 14C-labeled internal standards have demonstrated synthesis by the islets of the cyclooxygenase products prostaglandin E2, prostaglandin F2 alpha, thromboxane B2 and 12- hydroxyheptadecatrienoic acid as well as the lipoxygenase product 12-hydroxyeicosatetraenoic acid (12-HETE). Islet synthesis of these compounds was suppressed with appropriate inhibitors of arachidonate metabolism. Synthesis of the identified metabolites from endogenous arachidonate has also been quantitated with the use of deuterated internal standards, capillary column gas chromatographic analyses, and negative ion-chemical ionization mass spectrometric measurements. The relative abundances of metabolites derived from exogenous, radiolabeled arachidonate versus endogenous precursor differed considerably, and 12-HETE was by far the most abundant of these metabolites synthesized from endogenous arachidonate. Platelets contaminating the isolated islet preparations have been excluded as the source of the identified arachidonate metabolites. These studies establish that cells intrinsic to pancreatic islets synthesize a clearly characterized profile of arachidonate lipoxygenase and cyclooxygenase products. The sensitive and specific mass spectrometric methods for quantitation of these compounds permit detailed evaluation of their possible participation in insulin secretion from isolated islets.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Islotes Pancreáticos/enzimología , Lipooxigenasa/metabolismo , Prostaglandina-Endoperóxido Sintasas/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico , Animales , Ácido Araquidónico , Ácidos Araquidónicos/biosíntesis , Radioisótopos de Carbono , Cromatografía Líquida de Alta Presión , Dinoprostona , Cromatografía de Gases y Espectrometría de Masas , Leucotrieno B4/biosíntesis , Técnicas de Cultivo de Órganos , Prostaglandinas E/biosíntesis , Ratas , Tromboxano B2/biosíntesis , Tritio
9.
Biochim Biophys Acta ; 794(1): 125-36, 1984 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-6428455

RESUMEN

Isolated pancreatic islets from the rat incubated with 28 mM glucose have been found to secrete more insulin and to synthesize greater amounts of arachidonate lipoxygenase and cyclooxygenase products than islets incubated with 3 mM glucose. This effect was not apparent in studies examining metabolism of radiolabeled arachidonate and was revealed only when the metabolites were quantitated with mass spectrometric measurements. That the glucose-induced synthesis of arachidonate metabolites may participate in insulin secretion was suggested by studies with inhibitors of arachidonate metabolism. Eicosa 5,8,11,14 tetrynoic acid (ETYA) suppressed glucose-induced insulin secretion by 63-74% at a concentration (20 microM) which inhibited the synthesis of arachidonate lipoxygenase and cyclooxygenase products by 90%. Indomethacin (10 microM) completely prevented islet synthesis of cyclooxygenase products but did not influence glucose-induced insulin secretion. Although indomethacin did not inhibit the conversion of exogenous, 3H-labeled arachidonate to [3H]12-HETE, it did significantly inhibit (41-72%) the synthesis of 12-HETE from endogenous precursor. This is presumed to reflect indirect effects of indomethacin on hydrolysis of arachidonate from phospholipids, as recently reported in platelets. These studies constitute the first demonstration that glucose stimulates the synthesis of a lipoxygenase product (12-HETE) from endogenous arachidonate by isolated islets, and that suppression of 12-HETE synthesis with ETYA reduces glucose-induced insulin secretion from isolated islets.


Asunto(s)
Ácido 5,8,11,14-Eicosatetrainoico/farmacología , Ácidos Araquidónicos/metabolismo , Ácidos Grasos Insaturados/farmacología , Glucosa/farmacología , Indometacina/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico , Animales , Ácido Araquidónico , Ácidos Araquidónicos/biosíntesis , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Cinética , Lipooxigenasa/metabolismo , Técnicas de Cultivo de Órganos , Prostaglandinas/biosíntesis , Ratas
10.
Biochim Biophys Acta ; 879(3): 399-409, 1986 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-3535899

RESUMEN

Our recent findings indicate that glucose-induced insulin secretion from isolated pancreatic islets is temporally associated with accumulation of substantial amounts of free arachidonic acid and that arachidonate may serve as a second messenger for intracellular calcium mobilization in islets. In an effort to determine the source of this released arachidonate, the endogenous fatty acid composition of phospholipids from islets has been determined by thin-layer chromatographic separation of the phospholipids, methanolysis to the fatty acid methyl esters, and quantitative gas chromatographic analyses. The relative abundance of phospholipids in islets as judged by their fatty acid content was phosphatidylcholine (PC), 0.63; phosphatidylethanolamine (PE), 0.23; phosphatidylinositol (PI), 0.067; phosphatidylserine (PS), 0.049. Arachidonate constituted 17% of the total islet fatty acid content, and PC contained 43% of total islet arachidonate. Islets incubated with [3H]arachidonate in the presence of 28 mM D-glucose incorporated radiolabel into PC with a considerably higher specific activity than that of PE, PS or PI. The total fatty acid content of PC from islets incubated with 28 mM glucose for 30 min was significantly lower than that of islets incubated with 3 mM glucose, and smaller effects were observed with PE, PS and PI. The molar decrement in PC arachidonate was 3.2 pmol/islet under these conditions, which is sufficient to account for the previously observed accumulation of free arachidonate (2 pmol/islet). A sensitive method involving negative ion-chemical ionization-mass spectrometric analyses of the pentafluorobenzyl esters of fatty acids derived from trace amounts of lysophosphatidylcholine (lyso-PC) was developed, and glucose-stimulation was found to reduce islet lyso-PC content by about 10-fold. These findings indicate that the insulin secretagogue D-glucose induces phospholipid hydrolysis in islets and suggest that PC may be the major source of free arachidonate which accumulates in glucose-stimulated islets.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Ácidos Grasos/metabolismo , Glucosa/farmacología , Islotes Pancreáticos/metabolismo , Fosfolípidos/metabolismo , Animales , Células Cultivadas , Hidrólisis , Técnicas In Vitro , Islotes Pancreáticos/efectos de los fármacos , Cinética , Masculino , Ratas , Ratas Endogámicas
11.
Biochim Biophys Acta ; 835(1): 1-17, 1985 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-3924104

RESUMEN

Deuterium-labelled standards of four regionally isomeric epoxyeicosatrienoic acids (EETs) and their hydrolysis products, the dihydroxyeicosatrienoic acids (DHETs), have been prepared and analyzed by capillary column gas chromatography (GC)-negative ion (NI)-methane chemical ionization (MCI)-mass spectrometry (MS) as the pentafluorobenzyl esters. As little as 40 pg of these compounds were readily visualized by these methods, and the deuterium-labelled standards were used in a stable isotope dilution mass spectrometric assay which was linear from near the detection limit over several orders of magnitude. NADPH-dependent synthesis of both EETs and DHETs from arachidonate by hepatic microsomal cytochrome P-450-mono-oxygenase activity was demonstrable with these methods and was significantly suppressed by the compound BW755C (500 microM), but not by eicosa-5,8,11,14-tetraynoic acid (ETYA, 20 microM) or by nordihydroguaiaretic acid (NDGA, 50 microM). All three compounds suppress glucose-induced insulin secretion and 12-hydroxyeicosatetraenoic acid (12-HETE) synthesis by isolated pancreatic islets with similar concentration dependence. Microsomes derived from isolated pancreatic islets synthesized less than 3% of the EET and DHET compounds as a comparable amount of hepatic microsomes. Intact islets synthesized less than 3% by mass of the EET and DHET compounds compared to the mass of 12-HETE produced by the islets. Islets also failed to convert 3H-labelled arachidonate to 3H-labelled EETs or DHETs under conditions where conversion to [3H]12-HETE and to [3H]prostaglandin E2 (but not to [3H]leukotriene C4, D4, or E4) was clearly demonstrable. Neither exogenous EETs nor leukotriene C4 stimulated insulin secretion from the isolated islets or reversed the suppression of glucose-induced secretion by the lipoxygenase inhibitor BW755C. The cytochrome P-450-monooxygenase inhibitor, metyrapone (50 microM), did not influence insulin secretion from the isolated islets under conditions where the lipoxygenase inhibitor, NDGA, suppressed glucose-induced secretion. These observations argue against the recently suggested hypothesis that EETs derived from arachidonate by monooxygenase action participate in glucose-induced insulin secretion by isolated pancreatic islets.


Asunto(s)
Ácidos Araquidónicos/metabolismo , Islotes Pancreáticos/enzimología , Microsomas Hepáticos/enzimología , 4,5-dihidro-1-(3-(trifluorometil)fenil)-1H-pirazol-3-amina , Ácido 5,8,11,14-Eicosatetrainoico/metabolismo , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Araquidonato Lipooxigenasas , Ácido Araquidónico , Catecoles/farmacología , Sistema Enzimático del Citocromo P-450 , Cromatografía de Gases y Espectrometría de Masas , Ácidos Hidroxieicosatetraenoicos/metabolismo , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/ultraestructura , Lipooxigenasa/metabolismo , Masoprocol , Metirapona/farmacología , Microsomas/enzimología , NADP/metabolismo , Oxigenasas/metabolismo , Pirazoles/farmacología , Ratas , SRS-A/farmacología
12.
Biochim Biophys Acta ; 1001(1): 16-24, 1989 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-2492199

RESUMEN

Recent evidence indicates that the arachidonate metabolite 12-hydroxy-5,8,10,14-eicosatetraenoic acid (12-HETE) or its precursor may act as a second messenger in stimulus-response coupling in a variety of cells including Aplysia neurons, adrenal glomerulosa cells, and pancreatic islets. The compound 12(S)-HETE is generated from the precursor 12(S)-hydroperoxy-5,8,10,14-eicosatetraenoic acid (12(S)-HPETE), which is a product of the 12-lipoxygenase enzyme. Some cells have recently been found to produce the enantiomer 12(R)-HETE, apparently via a cytochrome P-450 monooxygenase, and the biologic actions of 12(R)-HETE and 12(S)-HETE differ. We have examined the stereochemistry of 12-HETE from isolated pancreatic islets both radiochemically and by a new mass spectrometric method capable of quantitating subnanogram amounts of 12-HETE stereoisomers. Endogenous 12-HETE from islets was found to be exclusively the S-isomer. D-Glucose stimulated both insulin secretion and islet accumulation of 12(S)-HETE but not of 12(R)-HETE. Pharmacologic inhibition of islet 12-HETE biosynthesis also suppressed glucose-induced insulin secretion. These findings suggest that islet 12-HETE is a product of a 12-lipoxygenase rather than of a cytochrome P-450 monooxygenase and further implicate 12-lipoxygenase products in stimulus-secretion coupling.


Asunto(s)
Araquidonato 12-Lipooxigenasa/metabolismo , Araquidonato Lipooxigenasas/metabolismo , Ácidos Araquidónicos/metabolismo , Islotes Pancreáticos/metabolismo , Ácido 12-Hidroxi-5,8,10,14-Eicosatetraenoico , Animales , Ácido Araquidónico , Cromatografía Líquida de Alta Presión , Glucosa/farmacología , Ácidos Hidroxieicosatetraenoicos/metabolismo , Ratas , Estereoisomerismo
13.
Biochim Biophys Acta ; 801(2): 197-205, 1984 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-6236851

RESUMEN

Studies were undertaken to determine whether the effect of alloxan to inactivate a membrane-bound calcium- and calmodulin-dependent protein kinase was specific for the pancreatic islets and whether inactivation of the kinase occurred also after injection of a diabetogenic dose of alloxan into rats. The effect of alloxan was also examined on similar particulate calcium- and calmodulin-dependent kinases present in two other secretory tissues, mammary acini and forebrain. Exposure of alloxan to cell-free preparations of all secretory tissues examined inhibited the calcium- and calmodulin-dependent kinase activities, suggesting that the specificity of alloxan action was not due to the presence in islets of a kinase uniquely sensitive to alloxan. To determine whether the selective effect of alloxan action was mediated at the cellular level, experiments were performed with alloxan presented to intact cells. Whereas alloxan exposure to viable cell preparations of islets and brain decreased the subsequently measured calcium- and calmodulin-dependent protein kinase activity, the activity measured in mammary acini exposed to these alloxan concentrations was unaffected. Injection (i.v.) of a diabetogenic dose of alloxan (50 mg/kg) produced an immediate (10 min) and selective inactivation of the calcium- and calmodulin-dependent protein kinase in pancreatic islets but had no effect on the similar kinases measured in brain and mammary acini. These results indicate that the unique sensitivity of islets to alloxan may result from the ability of alloxan to rapidly gain intracellular access and then inactivate this kinase activity. The selective effect of alloxan injection on this islet protein kinase is consistent with the hypothesis that inactivation of the kinase by alloxan is related to its diabetogenic effect in vivo.


Asunto(s)
Aloxano/farmacología , Encéfalo/enzimología , Islotes Pancreáticos/enzimología , Inhibidores de Proteínas Quinasas , Animales , Membrana Celular/enzimología , Diabetes Mellitus Experimental/enzimología , Islotes Pancreáticos/efectos de los fármacos , Cinética , Masculino , Glándulas Mamarias Animales/enzimología , Peso Molecular , Fenitoína/farmacología , Proteínas Quinasas/aislamiento & purificación , Ratas , Ratas Endogámicas , Estreptozocina/farmacología
14.
Biochim Biophys Acta ; 1361(3): 313-20, 1997 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-9375806

RESUMEN

The cytokine interleukin-1beta (IL-1beta) has been shown to inhibit insulin secretion and destroy pancreatic islets by a mechanism that involves the expression of inducible nitric oxide synthase (iNOS), and the production of nitric oxide (NO). Insulin containing beta-cells, selectively destroyed during the development of autoimmune diabetes, appear to be the islet cellular source of iNOS following treatment with IL-1beta. In this study we have evaluated the presence of type I IL-1 signaling receptors on purified pancreatic beta-cells. We show that the interleukin-1 receptor antagonist protein (IRAP) prevents IL-1beta-induced nitrite formation and IL-1beta-induced inhibition of insulin secretion by isolated islets and primary beta-cells purified by fluorescence-activated cell sorting (FACS). The protective effects of IRAP correlate with an inhibition of IL-1beta-induced iNOS expression by islets and FACS purified beta-cells. To provide direct evidence to support beta-cell expression of IL-1 type I signaling receptors, we show that antiserum specific for the type I IL-1 receptor neutralizes IL-1beta-induced nitrite formation by RINm5F cells, and that RINm5F cells express the type I IL-1 receptor at the protein level. Using reverse transcriptase-polymerase chain reaction (RT-PCR), the expression of type I IL-1 signaling receptors by FACS purified beta-cells and not alpha-cells is demonstrated. These results provide direct support for the expression of type I IL-1 receptors by primary pancreatic beta-cells, the cell type selectively destroyed during the development of autoimmune diabetes.


Asunto(s)
Islotes Pancreáticos/metabolismo , Receptores de Interleucina-1/metabolismo , Animales , Línea Celular , Citometría de Flujo , Insulina/metabolismo , Antagonistas de Insulina/farmacología , Secreción de Insulina , Proteína Antagonista del Receptor de Interleucina 1 , Masculino , Ratas , Ratas Sprague-Dawley , Sialoglicoproteínas/farmacología
15.
Biochim Biophys Acta ; 1300(1): 63-72, 1996 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-8608164

RESUMEN

Inflammatory cytokines may participate in the destruction of pancreatic islets during the pathogenesis of insulin-dependent diabetes mellitus, and the cytokine interleukin-1 (IL-1) strongly inhibits insulin secretion from rat pancreatic islets by a process which involves induction of expression of the inducible isoform of nitric oxide synthase and the overproduction of nitric oxide. The signaling events between IL-1 receptor occupancy and induction of nitric oxide synthase in rat islets involve activation of the transcriptional activator NFkappa B. Because sphingomyelin hydrolysis has been implicated as a signaling process both in NFkappa B activation and in IL-1 action in some cells, we have examined the potential involvement of sphingomyelin hydrolysis in the induction of islet nitric oxide overproduction by IL-1. Rat islet sphingomyelin pools were radiolabeled with [3H]choline, and sphingomyelin was then isolated by normal phase HPLC. Electrospray ionization-mass spectrometric analysis revealed islet sphingomyelin consists of at least 4 distinct molecular species, and the most abundant of them contained sphingosine as the long chain base and a residue of palmitic acid as the fatty acid substituent. Molecular species containing residues of stearic acid and arachidic acid were also observed. Neither interleukin-1 nor tumor necrosis factor-alpha was found to induce hydrolysis of islet sphingomyelin species, and neither an exogenous, cell-permeant ceramide species (N-acetyl-D-sphingosine) nor exogenous sphingomyelinase mimicked or potentiated the effect of IL-1 to increase rat islet nitric oxide generation, as reflected by nitrite production. Similar findings were obtained with RINm5F insulinoma cells and with mouse pancreatic islets. These findings provide the first information on the molecular species of sphingomyelin in pancreatic islets and suggest that sphingomyelin hydrolysis is not involved in the signaling pathway whereby IL-1 induces the overproduction of nitric oxide by pancreatic islets.


Asunto(s)
Interleucina-1/farmacología , Islotes Pancreáticos/efectos de los fármacos , Óxido Nítrico/metabolismo , Esfingomielinas/metabolismo , Animales , Células Cultivadas , Ceramidas/farmacología , Cromatografía Líquida de Alta Presión , Hidrólisis , Técnicas In Vitro , Islotes Pancreáticos/citología , Masculino , Espectrometría de Masas , Ratones , Ratas , Ratas Sprague-Dawley , Transducción de Señal , Factor de Necrosis Tumoral alfa/farmacología
16.
Diabetes ; 41(8): 897-903, 1992 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-1378415

RESUMEN

Cytokines have been implicated as immunological effector molecules that induce dysfunction and destruction of the pancreatic beta-cell. The mechanisms of cytokine action on the beta-cell are unknown; however, nitric oxide, resulting from cytokine-induced expression of nitric oxide synthase, has been implicated as the cellular effector molecule mediating beta-cell dysfunction. Nitric oxide is a free radical that targets intracellular iron-containing enzymes, which results in the loss of their function. The cytokine IL-1 beta induces the formation of nitric oxide in isolated rat islets and the insulinoma cell line, Rin-m5F. NMMA and NAME, both inhibitors of nitric oxide synthase, completely protect islets from the deleterious effects of IL-1 beta. These inhibitors are competitive in nature and inhibit both the cytokine-inducible and constitutive isoforms of nitric oxide synthase with nearly identical kinetics. This may preclude their use as therapeutic agents because of increases in blood pressure which result from the inhibition of constitutive nitric oxide synthase activity. Aminoguanidine, an inhibitor of nonenzymatic glycosylation of cellular and extracellular constituents associated with diabetic complications, recently has been reported to inhibit nitric oxide synthase. Aminoguanidine is approximately 40-fold more effective in inhibiting the inducible isoform of nitric oxide synthase, suggesting that aminoguanidine or analogues may serve as potential therapeutic agents to block diseases associated with nitric oxide production by the inducible isoform of nitric oxide synthase. In vivo administration of TNF IL-1 has been shown to induce anti-diabetogenic effects in the NOD mouse. This anti-diabetogenic effect of cytokines appears to conflict with evidence suggesting that cytokines mediate beta-cell dysfunction.(ABSTRACT TRUNCATED AT 250 WORDS)


Asunto(s)
Autoinmunidad/fisiología , Citocinas/fisiología , Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Óxido Nítrico/metabolismo , Aminoácido Oxidorreductasas/antagonistas & inhibidores , Animales , Muerte Celular/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Óxido Nítrico Sintasa
17.
Diabetes ; 30(4): 341-5, 1981 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7009279

RESUMEN

The present experiments indicate that ZnCl2 (0.015-0.50 mM) inhibits in a dose-dependent manner insulin secretion from isolated rat islets stimulated by D-glucose, L-leucine, and potassium. This inhibitory effect is partially reversed by washing and antagonized by high calcium concentrations in the medium. Zinc levels that inhibit insulin release do not affect 45calcium uptake, and zinc will not replace calcium in triggering insulin release. The conversion of 14C-D-glucose to 14CO2 by islets is not modified by zinc (0.12 mM or 0.50 mM) following either 2- or 0.5-h incubation periods, respectively. It is concluded that the inhibitory effect of zinc on insulin secretion may, in part, be mediated through interference with an intracellular function of calcium by the beta-cell.


Asunto(s)
Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Zinc/farmacología , Animales , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Glucosa , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Leucina , Potasio , Ratas
18.
Diabetes ; 24(9): 836-41, 1975 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-1098951

RESUMEN

The effects of cyproheptadine on basal and glucose-induced insulin release by isolated rat islets was studied by use of a perifusion system. A forty-five minute preincubation of islets with a medium containing both 34.3 mug./ml. (10(-4) M) cyproheptadine and 1.0 mg./ml. glucose completely abolished the biphasic pattern of increased insulin secretion normally obtained after islets are stimulated with a medium containing 3.0 mg./ml. glucose. In another series of experiments, similar results were obtained when the cyproheptadine and 3.0 mg./ml. glucose were presented together. Here, however, the inhibition of the first phase of insulin secretion did not achieve statistical significance and some recovery of the islets' secretory capacity was observed late during the second phase. In studies designed to investigate the influence of cyproheptadine on basal insulin secretion, no obvious effect was observed. These results are discussed in relation to the species-specific alterations in pancreatic beta-cell morphology that have been reported in rats after the oral administration of cyproheptadine.


Asunto(s)
Ciproheptadina/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Sinergismo Farmacológico , Glucosa/farmacología , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Masculino , Perfusión , Ratas
19.
Diabetes ; 27(2): 71-7, 1978 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-342321

RESUMEN

Isolated rat islets were maintained in a simple static incubation system and were exposed to alloxan for a period of five minutes. Alloxan inhibited subsequent glucose-induced insulin release in a dose-dependent manner at 37 degrees C., with 650 muM alloxan producing 94 per cent inhibition of insulin release. Barbituric acid, a compound structurally related to alloxan, provided complete protection (at 37 degrees C.) against this inhibition of insulin release when present during the alloxan exposure. At 23 degrees C., barbituric acid was shown to be absent from the intracellular space of the islet yet still protected completely against alloxan inhibition of insulin release. Thus, barbituric acid apparently provided protection against alloxan in the extracellular medium. By fluorometric and chromatographic analyses, it was determined that barbituric acid reacted rapidly with alloxan to produce a new compound. These findings indicate that barbituric acid protected against alloxan by a chemical reaction in the medium.


Asunto(s)
Aloxano/farmacología , Barbitúricos/farmacología , Glucosa/farmacología , Insulina/metabolismo , Islotes Pancreáticos/metabolismo , Animales , Secreción de Insulina , Islotes Pancreáticos/efectos de los fármacos , Cinética , Ratas , Factores de Tiempo
20.
Diabetes ; 50(2): 353-60, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11272147

RESUMEN

Recent findings have demonstrated that the branched-chain amino acid leucine can activate the translational regulators, phosphorylated heat- and acid-stable protein regulated by insulin (PHAS-I) and p70 S6 kinase (p70S6k), in an insulin-independent and rapamycin-sensitive manner through mammalian target of rapamycin (mTOR), although the mechanism for this activation is undefined. It has been previously established that leucine-induced insulin secretion by beta-cells involves increased mitochondrial metabolism by oxidative decarboxylation and allosteric activation of glutamate dehydrogenase (GDH). We now show that these same intramitochondrial events that generate signals for leucine-induced insulin exocytosis are required to activate the mTOR mitogenic signaling pathway by beta-cells. Thus, a minimal model consisting of leucine and glutamine as substrates for oxidative decarboxylation and an activator of GDH, respectively, confirmed the requirement for these two metabolic components and mimicked closely the synergistic interactions achieved by a complete complement of amino acids to activate p70s6k in a rapamycin-sensitive manner. Studies using various leucine analogs also confirmed the close association of mitochondrial metabolism and the ability of leucine analogs to activate p70s6k. Furthermore, selective inhibitors of mitochondrial function blocked this activation in a reversible manner, which was not associated with a global reduction in ATP levels. These findings indicate that leucine at physiological concentrations stimulates p70s6k phosphorylation via the mTOR pathway, in part, by serving both as a mitochondrial fuel and an allosteric activator of GDH. Leucine-mediated activation of protein translation through mTOR may contribute to enhanced beta-cell function by stimulating growth-related protein synthesis and proliferation associated with the maintenance of beta-cell mass.


Asunto(s)
Islotes Pancreáticos/fisiología , Leucina/fisiología , Fosfotransferasas (Aceptor de Grupo Alcohol)/fisiología , Biosíntesis de Proteínas/fisiología , Proteínas Quinasas , Regulación Alostérica , Aminoácidos Cíclicos/farmacología , Línea Celular , Descarboxilación , Activación Enzimática , Glutamato Deshidrogenasa/metabolismo , Ácido Glutámico/fisiología , Islotes Pancreáticos/citología , Isoleucina/farmacología , Leucina/metabolismo , Leucina/farmacología , Mitocondrias/fisiología , Modelos Biológicos , Oxidación-Reducción , Fosforilación , Proteínas Quinasas S6 Ribosómicas/metabolismo , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR , Valina/farmacología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda