Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Endocrinol Invest ; 47(1): 245-253, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37354249

RESUMO

PURPOSE: There is little information on factors that influence the glycemic variability (GV) during the nocturnal and diurnal periods. We aimed to examine the relationship between clinical factors and GV during these two periods. METHODS: This cross-sectional study included 134 patients with type 2 diabetes. 24-h changes in blood glucose were recorded by a continuous glucose monitoring system. Nocturnal and diurnal GV were assessed by standard deviation of blood glucose (SDBG), coefficient of variation (CV), and mean amplitude of glycemic excursions (MAGE), respectively. Robust regression analyses were performed to identify the factors associated with GV. Restricted cubic splines were used to determine dose-response relationship. RESULTS: During the nocturnal period, age and glycemic level at 12:00 A.M. were positively associated with GV, whereas alanine aminotransferase was negatively associated with GV. During the diurnal period, homeostatic model assessment 2-insulin sensitivity (HOMA2-S) was positively associated with GV, whereas insulin secretion-sensitivity index-2 (ISSI2) was negatively associated with GV. Additionally, we found a J-shape association between the glycemic level at 12:00 A.M. and MAGE, with 9.0 mmol/L blood glucose level as a cutoff point. Similar nonlinear associations were found between ISSI2 and SDBG, and between ISSI2 and MAGE, with ISSI2 value of 175 as a cutoff point. CONCLUSION: Factors associated with GV were different between nocturnal and diurnal periods. The cutoff points we found in this study may provide the therapeutic targets for beta-cell function and pre-sleep glycemic level in clinical practice.


Assuntos
Diabetes Mellitus Tipo 2 , Resistência à Insulina , Humanos , Diabetes Mellitus Tipo 2/tratamento farmacológico , Glicemia/análise , Estudos Transversais , Automonitorização da Glicemia , Resistência à Insulina/fisiologia
2.
Diabetologia ; 56(2): 359-69, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23132338

RESUMO

AIMS/HYPOTHESIS: The molecular basis of the exocytosis of secretory insulin-containing granules (SGs) during biphasic glucose-stimulated insulin secretion (GSIS) from pancreatic beta cells remains unclear. Syntaxin (SYN)-1A and SYN-4 have been shown to mediate insulin exocytosis. The insulin-secretory function of SYN-3, which is particularly abundant in SGs, is unclear. METHODS: Mouse pancreatic islets and INS-1 cells were treated with adenovirus carrying Syn-3 (also known as Stx3) or small interfering RNA targeting Syn-3 in order to examine insulin secretion by radioimmunoassay. The localisation and distribution of insulin granules were examined by confocal and electron microscopy. Dynamic single-granule fusion events were assessed using total internal reflection fluorescence microscopy (TIRFM). RESULTS: Depletion of endogenous SYN-3 inhibited insulin release. TIRFM showed no change in the number or fusion competence of previously docked SGs but, instead, a marked reduction in the recruitment of newcomer SGs and their subsequent exocytotic fusion during biphasic GSIS. Conversely, overexpression of Syn-3 enhanced both phases of GSIS, owing to the increase in newcomer SGs and, remarkably, to increased SG-SG fusion, which was confirmed by electron microscopy. CONCLUSIONS/INTERPRETATION: In insulin secretion, SYN-3 plays a role in the mediation of newcomer SG exocytosis and SG-SG fusion that contributes to biphasic GSIS.


Assuntos
Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas Qa-SNARE/metabolismo , Animais , Western Blotting , Linhagem Celular , Exocitose/genética , Humanos , Secreção de Insulina , Camundongos , Microscopia Confocal , Microscopia Eletrônica , Microscopia de Fluorescência , Proteínas Qa-SNARE/genética , RNA Interferente Pequeno , Radioimunoensaio
3.
Diabetologia ; 55(4): 877-80, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22307686

RESUMO

According to our current understanding of insulin exocytosis, insulin granules dock on the plasma membrane, undergo priming and then wait for calcium-triggered fusion. In this issue of Diabetologia, Hoppa et al (doi 10.1007/s00125-011-2400-5 ) report that cholinergic stimulation induces granule-granule, or multivesicular, fusion to effect more efficient insulin release. Other exocytotic modes of insulin secretion, particularly those induced by incretin stimulation, include orderly granule fusion with granules already fused with the plasma membrane, called sequential exocytosis, and recruitment of newcomer granules to fuse with plasma membrane with minimal time for docking and priming. The molecular machineries that mediate these distinct exocytotic modes of granule-granule fusion and newcomer granules remain undefined, but they could be therapeutically targeted to couple to cholinergic and incretin stimulation to rescue the deficient glucose-stimulated insulin secretion in diabetes.


Assuntos
Exocitose/fisiologia , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Vesículas Secretórias/metabolismo , Animais , Secreção de Insulina
4.
Diabetologia ; 55(10): 2693-2702, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22814762

RESUMO

AIMS/HYPOTHESIS: We have previously reported that the haplodeficient Munc13-1(+/-) mouse exhibits impaired biphasic glucose-stimulated insulin secretion (GSIS), causing glucose intolerance mimicking type 2 diabetes. Glucagon-like peptide-1 (GLP-1) can bypass these insulin-secretory defects in type 2 diabetes, but the mechanism of exocytotic events mediated by GLP-1 in rescuing insulin secretion is unclear. METHODS: The total internal reflection fluorescence microscopy (TIRFM) technique was used to examine single insulin granule fusion events in mouse islet beta cells. RESULTS: There was no difference in the density of docked granules in the resting state between Munc13-1(+/+) and Munc13-1(+/-) mouse islet beta cells. While exocytosis of previously docked granules in Munc13-1(+/-) beta cells is reduced during high-K(+) stimulation as expected, we now find a reduction in additional exocytosis events that account for the major portion of GSIS, namely two types of newcomer granules, one which has a short docking time (short-dock) and another undergoing no docking before exocytosis (no-dock). As mammalian homologue of Caenorhabditis elegans unc-13-1 (Munc13-1) is a phorbol ester substrate, phorbol ester could partially rescue biphasic GSIS in Munc13-1-deficient beta cells by enhancing recruitment of short-dock newcomer granules for exocytosis. The more effective rescue of biphasic GSIS by GLP-1 than by phorbol was due to increased recruitment of both short-dock and no-dock newcomer granules. CONCLUSIONS/INTERPRETATION: Phorbol ester and GLP-1 potentiation of biphasic GSIS are brought about by recruitment of distinct populations of newcomer granules for exocytosis, which may be mediated by Munc13-1 interaction with syntaxin-SNARE complexes other than that formed by syntaxin-1A.


Assuntos
Exocitose/fisiologia , Glucose/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Animais , Exocitose/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/fisiologia , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Ésteres de Forbol/farmacologia , Potássio/farmacologia , Proteínas Qa-SNARE/fisiologia , Proteínas SNARE/fisiologia , Sintaxina 1/fisiologia
5.
Diabetologia ; 55(6): 1709-20, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22411134

RESUMO

AIMS/HYPOTHESIS: It is thought that the voltage-dependent potassium channel subunit Kv2.1 (Kv2.1) regulates insulin secretion by controlling beta cell electrical excitability. However, this role of Kv2.1 in human insulin secretion has been questioned. Interestingly, Kv2.1 can also regulate exocytosis through direct interaction of its C-terminus with the soluble NSF attachment receptor (SNARE) protein, syntaxin 1A. We hypothesised that this interaction mediates insulin secretion independently of Kv2.1 electrical function. METHODS: Wild-type Kv2.1 or mutants lacking electrical function and syntaxin 1A binding were studied in rodent and human beta cells, and in INS-1 cells. Small intracellular fragments of the channel were used to disrupt native Kv2.1-syntaxin 1A complexes. Single-cell exocytosis and ion channel currents were monitored by patch-clamp electrophysiology. Interaction between Kv2.1, syntaxin 1A and other SNARE proteins was probed by immunoprecipitation. Whole-islet Ca(2+)-responses were monitored by ratiometric Fura red fluorescence and insulin secretion was measured. RESULTS: Upregulation of Kv2.1 directly augmented beta cell exocytosis. This happened independently of channel electrical function, but was dependent on the Kv2.1 C-terminal syntaxin 1A-binding domain. Intracellular fragments of the Kv2.1 C-terminus disrupted native Kv2.1-syntaxin 1A interaction and impaired glucose-stimulated insulin secretion. This was not due to altered ion channel activity or impaired Ca(2+)-responses to glucose, but to reduced SNARE complex formation and Ca(2+)-dependent exocytosis. CONCLUSIONS/INTERPRETATION: Direct interaction between syntaxin 1A and the Kv2.1 C-terminus is required for efficient insulin exocytosis and glucose-stimulated insulin secretion. This demonstrates that native Kv2.1-syntaxin 1A interaction plays a key role in human insulin secretion, which is separate from the channel's electrical function.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Canais de Potássio Shab/metabolismo , Animais , Linhagem Celular Tumoral , Células Cultivadas , Eletrofisiologia , Humanos , Immunoblotting , Imunoprecipitação , Secreção de Insulina , Camundongos , Ligação Proteica , Ratos , Canais de Potássio Shab/genética , Sintaxina 1/metabolismo
6.
Diabetologia ; 53(8): 1656-68, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20424817

RESUMO

AIMS/HYPOTHESIS: Zinc is highly concentrated in pancreatic beta cells, is critical for normal insulin storage and may regulate glucagon secretion from alpha cells. Zinc transport family member 8 (ZnT8) is a zinc efflux transporter that is highly abundant in beta cells. Polymorphisms of ZnT8 (also known as SLC30A8) gene in man are associated with increased risk of type 2 diabetes. While global Znt8 knockout (Znt8KO) mice have been characterised, ZnT8 is also present in other islet cell types and extra-pancreatic tissues. Therefore, it is important to find ways of understanding the role of ZnT8 in beta and alpha cells without the difficulties caused by the confounding effects of ZnT8 in these other tissues. METHODS: We generated mice with beta cell-specific (Znt8BKO) and alpha cell-specific (Znt8AKO) knockout of Znt8, and performed in vivo and in vitro characterisation of the phenotypes to determine the functional and anatomical impact of ZnT8 in these cells. Thus we assessed zinc accumulation, insulin granule morphology, insulin biosynthesis and secretion, and glucose homeostasis. RESULTS: Znt8BKO mice are glucose-intolerant, have reduced beta cell zinc accumulation and atypical insulin granules. They also display reduced first-phase glucose-stimulated insulin secretion, reduced insulin processing enzyme transcripts and increased proinsulin levels. In contrast, Znt8AKO mice show no evident abnormalities in plasma glucagon and glucose homeostasis. CONCLUSIONS/INTERPRETATION: This is the first report of specific beta and alpha cell deletion of Znt8. Our data indicate that while, under the conditions studied, ZnT8 is absolutely essential for proper beta cell function, it is largely dispensable for alpha cell function.


Assuntos
Proteínas de Transporte de Cátions/genética , Glucose/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Zinco/metabolismo , Análise de Variância , Animais , Western Blotting , Proteínas de Transporte de Cátions/metabolismo , Grânulos Citoplasmáticos/genética , Grânulos Citoplasmáticos/metabolismo , Células Secretoras de Glucagon/metabolismo , Imuno-Histoquímica , Insulina/genética , Secreção de Insulina , Camundongos , Camundongos Knockout , Microscopia Confocal , Microscopia Imunoeletrônica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transportador 8 de Zinco
7.
Diabetologia ; 51(12): 2290-8, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18850083

RESUMO

AIMS/HYPOTHESIS: The hyperpolarisation-activated cyclic nucleotide-gated (HCN) channels, discovered initially in cardiac and neuronal cells, mediate the inward pacemaker current (I (f) or I (h)). Recently, we have demonstrated the presence of HCN channels in pancreatic beta cells. Here, we aim to examine the presence and function of HCN channels in glucagon-secreting alpha cells. METHODS: RT-PCR and immunocytochemistry were used to examine the presence of HCN channels in alpha cells. Whole-cell patch-clamp, calcium imaging and glucagon secretion experiments were performed to explore the function of HCN channels in alpha cells. RESULTS: HCN transcripts and proteins were detected in alpha-TC6 cells and dispersed rat alpha cells. Patch-clamp recording showed hyperpolarisation-activated currents in alpha-TC6 cells, which could be blocked by HCN channel inhibitor ZD7288. Glucagon secretion RIA studies demonstrated that at both low and high glucose concentrations (2 and 20 mmol/l), ZD7288 significantly enhanced glucagon secretion in alpha-TC6 and IN-R1-G9 cell lines. Conversely, activation of HCN channels by lamotrigine significantly suppressed glucagon secretion at the low glucose concentration. Calcium imaging studies showed that blockade of HCN channels by ZD7288 significantly increased intracellular calcium in alpha-TC6 cells, while lamotrigine or the Na(+) channel blocker tetrodotoxin suppressed the effect of ZD7288 on intracellular calcium. Furthermore, we found the HCN channel inhibitors ZD7288 and cilobradine both significantly increased glucagon secretion from rat islets. CONCLUSIONS/INTERPRETATION: These results suggest a potential role for HCN channels in regulation of glucagon secretion via modulating Ca(2+) and Na(+) channel activities.


Assuntos
Canais de Cátion Regulados por Nucleotídeos Cíclicos/metabolismo , Ilhotas Pancreáticas/metabolismo , Animais , Cálcio/metabolismo , Células Cultivadas , Canais de Cátion Regulados por Nucleotídeos Cíclicos/genética , Eletrofisiologia , Regulação da Expressão Gênica , Glucagon/metabolismo , Masculino , Técnicas de Patch-Clamp , Ratos , Ratos Wistar , Sódio/metabolismo
8.
J Clin Invest ; 83(1): 321-5, 1989 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2910915

RESUMO

The molecular events that mediate cholecystokinin (CCK)-stimulated pancreatic secretion are not well defined because of the complex receptor-binding and concentration-response characteristics of this hormone. Functional models of receptor occupancy initiating the cascade leading to secretion have been complicated by the inhibition of secretion effected by supramaximal concentrations of CCK. Recent report of a CCK analogue that does not exhibit supramaximal inhibition led us to synthesize a similar analogue that could also be radiolabeled for studies of receptor binding and affinity labeling, and for studies of second messenger activity. This probe, D-Tyr-Gly-[(Nle28,31)CCK-26-32]-phenethyl ester, was a fully efficacious secretagogue with no supramaximal inhibition, and, unlike native hormone, bound to a single class of sites present on both acini and membranes. Occupation of this site correlated well with stimulation of secretion. Evidence that this was indeed a CCK-binding site were the abilities of CCK and the antagonist L-364, 718 to inhibit binding of this analogue. Affinity labeling confirmed the identity of the site mediating secretory stimulation as a Mr = 85,000-95,000 protein. Whereas the nonhydrolyzable guanosine triphosphate analogue, 5'-guanylyl-imidodiphosphate, was a potent inhibitor of CCK binding, it had no effect on binding of this secretagogue, suggesting that a novel cascade not involving a guanine nucleotide-binding protein mediates CCK stimulation of pancreatic secretion.


Assuntos
Colecistocinina/farmacologia , Pâncreas/enzimologia , Animais , Relação Dose-Resposta a Droga , Masculino , Peso Molecular , Ratos , Ratos Endogâmicos , Sistemas do Segundo Mensageiro , Sincalida/farmacologia
9.
J Clin Invest ; 108(11): 1597-611, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11733555

RESUMO

Exocytosis at the apical surface of pancreatic acinar cells occurs in the presence of physiological concentrations of cholecystokinin (CCK) but is inhibited at high concentrations. Here we show that Munc18c is localized predominantly to the basal membranes of acinar cells. Supramaximal but not submaximal CCK stimulation caused Munc18c to dissociate from the plasma membrane, and this displacement was blocked by protein kinase C (PKC) inhibitors. Conversely, whereas the CCK analog CCK-OPE alone failed to displace Munc18c from the membrane, this agent caused Munc18c displacement following minimal PKC activation. To determine the physiological significance of this displacement, we used the fluorescent dye FM1-43 to visualize individual exocytosis events in real-time from rat acinar cells in culture. We showed that supramaximal CCK inhibition of secretion resulted from impaired apical secretion and a redirection of exocytic events to restricted basal membrane sites. In contrast, CCK-OPE evoked apical exocytosis and could only induce basolateral exocytosis following activation of PKC. Infusion of supraphysiological concentrations of CCK in rats, a treatment that induced tissue changes reminiscent of mild acute pancreatitis, likewise resulted in rapid displacement of Munc18c from the basal membrane in vivo.


Assuntos
Colecistocinina/farmacologia , Exocitose , Proteínas do Tecido Nervoso , Pâncreas/metabolismo , Proteínas/metabolismo , Proteínas de Transporte Vesicular , Animais , Membrana Celular/metabolismo , Colecistocinina/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Munc18 , Pancreatite/etiologia , Proteína Quinase C/fisiologia , Proteínas/análise , Ratos , Proteínas SNARE , Acetato de Tetradecanoilforbol/farmacologia
10.
Mol Biol Cell ; 7(12): 2019-27, 1996 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8970162

RESUMO

Syntaxins are cytoplasmically oriented integral membrane soluble NEM-sensitive factor receptors (SNAREs; soluble NEM-sensitive factor attachment protein receptors) thought to serve as targets for the assembly of protein complexes important in regulating membrane fusion. The SNARE hypothesis predicts that the fidelity of vesicle traffic is controlled in part by the correct recognition of vesicle SNAREs with their cognate target SNARE partner. Here, we show that in the exocrine acinar cell of the pancreas, multiple syntaxin isoforms are expressed and that they appear to reside in distinct membrane compartments. Syntaxin 2 is restricted to the apical plasma membrane whereas syntaxin 4 is found most abundantly on the basolateral membranes. Surprisingly, syntaxin 3 was found to be localized to a vesicular compartment, the zymogen granule membrane. In addition, we show that these proteins are capable of specific interaction with vesicle SNARE proteins. Their nonoverlapping locations support the general principle of the SNARE hypothesis and provide new insights into the mechanisms of polarized secretion in epithelial cells.


Assuntos
Proteínas de Membrana/metabolismo , Pâncreas/metabolismo , Animais , Anticorpos Monoclonais/análise , Extratos Celulares , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Pâncreas/citologia , Proteínas Qa-SNARE , Coelhos , Ratos , Ratos Sprague-Dawley
11.
Diabetes ; 48(5): 997-1005, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10331403

RESUMO

In normal isolated beta-cells, the response to glucose is heterogeneous and characterized by an increasing number of secretory cells as glucose concentration rises (Pipeleers DG, Kiekens R, Ling Z, Wilikens A, Schuit F: Physiologic relevance of heterogeneity in the pancreatic beta-cell population. Diabetologia 37 (Suppl. 2):S57-S64, 1994). We hypothesized that fasting hyperinsulinemia in obesity might be explained by altered beta-cell heterogeneity of signal transduction mechanisms, possibly involving exocytotic proteins. Insulin secretion from individual beta-cells sorted according to the size of the islet donor (<125 microm, >250 microm, and intermediate diameter) was measured by reverse hemolytic plaque assay. Beta-cells from fa/fa rats were hypertrophied 25-40%, independent of donor islet size. This was accompanied by an increased proportion of secretory cells (recruitment) at 5.5-11.0 mmol/l glucose, increased secretion per cell at 2.8 mmol/l glucose, and decreased insulin content after acute glucose exposure without an increase in secretion per cell. Decreased expression of exocytotic (soluble N-ethylmaleimide-sensitive fusion protein receptor [SNARE]) proteins, vesicle-associated membrane protein isoform 2 (VAMP-2), synaptosomal protein of 25 kDa (SNAP-25), and syntaxin-1 and -2 in fa/fa beta-cells may contribute to the failure to sustain excessive plaque size at higher glucose concentrations. Fasting hyperinsulinemia may be maintained by increased recruitment and an exaggerated secretory response in all fa-derived islet populations. Glucose regulates beta-cell responsiveness in the short term, and these effects may involve altered expression of SNARE proteins.


Assuntos
Glucose/farmacologia , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/patologia , Proteínas de Membrana/análise , Proteínas de Transporte Vesicular , Animais , Glicemia/metabolismo , Exocitose , Feminino , Hipertrofia , Insulina/sangue , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Masculino , Proteínas do Tecido Nervoso/análise , Obesidade/patologia , Obesidade/fisiopatologia , Proteínas Qa-SNARE , Proteínas R-SNARE , Ratos , Ratos Zucker , Proteínas SNARE , Proteína 25 Associada a Sinaptossoma , Sintaxina 1
12.
Mol Endocrinol ; 13(8): 1305-17, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10446905

RESUMO

Glucagon-like peptide-1 (GLP-1) is an insulinotropic hormone with powerful antidiabetogenic effects that are thought to be mediated by adenylyl cyclase (AC). Recently, we generated two GLP-1 receptor mutant isoforms (IC3-1 and DM-1) that displayed efficient ligand binding and the ability to promote Ca2+ mobilization from intracellular stores but lacked the ability to couple to AC. In the present study, the wild-type rat GLP-1 receptor (WT-GLP-1 R) or the IC3-1 and DM-1 mutant forms were expressed for the first time in the insulin-producing HIT-T15 cells. Only cells expressing WT-GLP-1 R displayed dramatically elevated GLP-1-induced cAMP responses and elevated insulin secretion. The increase in GLP-1-stimulated secretion in cells expressing WT-GLP-1 R, however, was not accompanied by differences in glucose-stimulated insulin release. Prolonged exposure to GLP-1 (10 nM, 17 h), not only led to an increase in insulin secretion but also increased insulin mRNA levels, but only in cells expressing the WT-GLP-1 R and not the mutant isoforms. Electrophysiological analyses revealed that GLP-1 application enhanced L-type voltage-dependent Ca2+ channel (VDCC) currents > 2-fold and caused a positive shift in VDCC voltage-dependent inactivation in WT-GLP-1R cells only, not control or mutant (DM-1) cells. This action on the Ca2+ current was further enhanced by the VDCC agonist, BAYK8644, suggesting GLP-1 acts via a distinct mechanism dependent on cAMP. These studies demonstrate that the GLP-1 receptor efficiently couples to AC to stimulate insulin secretion and that receptors lacking critical residues in the proximal region of the third intracellular loop can effectively uncouple the receptor from cAMP production, VDCC activity, insulin secretion, and insulin biosynthesis.


Assuntos
Glucagon/farmacologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Mutagênese , Fragmentos de Peptídeos/farmacologia , Precursores de Proteínas/farmacologia , Receptores de Glucagon/genética , Receptores de Glucagon/fisiologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Animais , Bário/metabolismo , Agonistas dos Canais de Cálcio/farmacologia , Linhagem Celular , AMP Cíclico/metabolismo , Condutividade Elétrica , Deleção de Genes , Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Glucose/farmacologia , Humanos , Insulina/biossíntese , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ratos , Relação Estrutura-Atividade , Transfecção
13.
Mol Endocrinol ; 12(7): 1060-70, 1998 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9658409

RESUMO

We and others have previously shown that insulin-secreting cells of the pancreas express high levels of SNAP-25 (synaptosomal-associated protein of 25 kDa), a 206-amino acid t-SNARE (target soluble N-ethylmaleimide-sensitive factor attachment protein receptors) implicated in synaptic vesicle exocytosis. In the present study, we show that SNAP-25 is required for insulin secretion by transient transfection of Botulinum Neurotoxin A (BoNT/A) into insulin-secreting HIT-T15 cells. Transient expression of BoNT/A cleaved the endogenous as well as overexpressed SNAP-25 proteins and caused significant reductions in K+ and glucose-evoked secretion of insulin. To determine whether the inhibition of release was due to the depletion of functional SNAP-25 or the accumulation of proteolytic by-products, we transfected cells with SNAP-25 proteins from which the C-terminal nine amino acids had been deleted to mimic the effects of the toxin. This modified SNAP-25 (amino acids 1-197) remained bound to the plasma membrane but was as effective as the toxin at inhibiting insulin secretion. Microfluorimetry revealed that the inhibition of secretion was due neither to changes in basal cytosolic Ca2+ levels nor in Ca2+ influx evoked by K(+)-mediated plasma membrane depolarization. Electron microscopy revealed that cells transfected with either BoNT/A or truncated SNAP-25 contained significantly higher numbers of insulin granules, many of which clustered close to the plasma membrane. Together, these results demonstrate that functional SNAP-25 proteins are required for insulin secretion and suggest that the inhibitory action of BoNT/A toxin on insulin secretion is in part caused by the production of the plasma membrane-bound cleavage product, which itself interferes with insulin granule docking and fusion.


Assuntos
Toxinas Botulínicas Tipo A/farmacologia , Insulina/metabolismo , Insulinoma/metabolismo , Proteínas de Membrana , Proteínas do Tecido Nervoso/farmacologia , Neoplasias Pancreáticas/metabolismo , Toxinas Botulínicas Tipo A/genética , Toxinas Botulínicas Tipo A/metabolismo , Cálcio/metabolismo , Membrana Celular/fisiologia , Grânulos Citoplasmáticos/ultraestrutura , Eletrofisiologia , Expressão Gênica , Glucose/farmacologia , Secreção de Insulina , Insulinoma/ultraestrutura , Microscopia Eletrônica , Mutagênese , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neoplasias Pancreáticas/ultraestrutura , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Potássio/farmacologia , Proteína 25 Associada a Sinaptossoma , Transfecção , Células Tumorais Cultivadas
14.
Mol Endocrinol ; 15(8): 1423-35, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11463864

RESUMO

In pancreatic beta-cells, voltage-dependent K(+) (Kv) channels are potential mediators of repolarization, closure of Ca(2+) channels, and limitation of insulin secretion. The specific Kv channels expressed in beta-cells and their contribution to the delayed rectifier current and regulation of insulin secretion in these cells are unclear. High-level protein expression and mRNA transcripts for Kv1.4, 1.6, and 2.1 were detected in rat islets and insulinoma cells. Inhibition of these channels with tetraethylammonium decreased I(DR) by approximately 85% and enhanced glucose-stimulated insulin secretion by 2- to 4-fold. Adenovirus-mediated expression of a C-terminal truncated Kv2.1 subunit, specifically eliminating Kv2 family currents, reduced delayed rectifier currents in these cells by 60-70% and enhanced glucose-stimulated insulin secretion from rat islets by 60%. Expression of a C-terminal truncated Kv1.4 subunit, abolishing Kv1 channel family currents, reduced delayed rectifier currents by approximately 25% and enhanced glucose-stimulated insulin secretion from rat islets by 40%. This study establishes that Kv2 and 1 channel homologs mediate the majority of repolarizing delayed rectifier current in rat beta-cells and that antagonism of Kv2.1 may prove to be a novel glucose-dependent therapeutic treatment for type 2 diabetes.


Assuntos
Insulina/metabolismo , Ilhotas Pancreáticas/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana , Canais de Potássio/fisiologia , 1-Metil-3-Isobutilxantina/farmacologia , Adenoviridae/genética , Animais , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Canais de Potássio de Retificação Tardia , Condutividade Elétrica , Deleção de Genes , Expressão Gênica , Vetores Genéticos , Glucose/farmacologia , Glibureto/farmacologia , Proteínas de Fluorescência Verde , Secreção de Insulina , Insulinoma/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Canal de Potássio Kv1.4 , Proteínas Luminescentes/genética , Neoplasias Pancreáticas/metabolismo , Bloqueadores dos Canais de Potássio , Canais de Potássio/genética , RNA Mensageiro/análise , Ratos , Proteínas Recombinantes , Canais de Potássio Shab , Tetraetilamônio/farmacologia , Transfecção
15.
Endocrinology ; 137(4): 1340-8, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8625909

RESUMO

Pancreatic beta cells and cell lines were used in the present study to test the hypothesis that the molecular mechanisms controlling exocytosis from neuronal cells may be used by the beta cell to regulate insulin secretion. Using specific antisera raised against an array of synaptic proteins (SNAREs) implicated in the control of synaptic vesicle fusion and exocytosis, we have identified the expression of several SNAREs in the islet beta cell lines, beta TC6-f7 and HIT-T15, as well as in pancreatic islets. The v-SNARE vesicle-associated membrane protein (VAMP)-2 but not VAMP-1 immunoreactive proteins were detected in beta TC6-f7 and HIT-T15 cells and pancreatic islets. In these islet-derived cell lines, this 18-kDa protein comigrated with rat brain synaptic vesicle VAMP-2, which was cleaved by Tetanus toxin (TeTx). Immunofluorescence confocal microscopy and electron microscopy localized the VAMP-2 to the cytoplasmic side of insulin containing secretory granule membrane. In streptolysin O permeabilized HIT-T15 cells, TeTx inhibited Ca2+-evoked insulin release by 83 +/- 4.3%, which correlated well to the cleavage of VAMP-2. The beta cell lines were also shown to express a second vesicle (v)-SNARE, cellubrevin. The proposed neuronal target (t)-membrane SNAREs, SNAP-25, and syntaxin isoforms 1-4 were also detected by Western blotting. The beta cell 25-kDa SNAP-25 protein and syntaxin isoforms 1-3 were specifically cleaved by botulinum A and C toxins, respectively, as observed with the brain isoforms. These potential t-SNARES were localized by immunofluorescence microscopy primarily to the plasma membrane in beta cell lines as well as in islet beta cells. To determine the specific identity of the immunoreactive syntaxin-2 and -3 isoforms and to explore the possibility that these beta cells express the putative Ca2+-sensing molecule synaptotagmin III, RT-PCR was performed on the beta cell lines. These studies confirmed that betaTC6-F7 cells express syntaxin-2 isoforms, 2 and 2', but not 2'' and express syntaxin-3. They further demonstrate the expression of synaptotagmin III. DNA sequence analysis revealed that rat and mouse beta cell syntaxins 2, 2' and synaptotagmin III are highly conserved at the nucleotide and predicted amino acid levels (95-98%). The presence of VAMP-2, nSec/Munc-18, SNAP-25 and syntaxin family of proteins, along with synaptotagmin III in the islet cells and in beta cell lines provide evidence that neurons and beta cells share similar molecular mechanisms for Ca2+-regulated exocytosis. The inhibition of Ca2+-evoked insulin secretion by the proteolytic cleavage of HIT-T15 cell VAMP-2 supports the hypothesis that these proteins play an integral role in the control of insulin exocytosis.


Assuntos
Proteínas de Ligação ao Cálcio , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas de Transporte Vesicular , Animais , Sequência de Bases , Toxinas Botulínicas/farmacologia , Linhagem Celular , Imuno-Histoquímica , Isomerismo , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Sondas de Oligonucleotídeos/genética , Proteínas Qa-SNARE , Proteínas R-SNARE , Ratos , Ratos Sprague-Dawley , Proteínas SNARE , Sinaptotagminas , Tetrodotoxina/farmacologia
16.
FEBS Lett ; 414(2): 298-302, 1997 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-9315706

RESUMO

The SNARE hypothesis proposes that specificity of exocytosis is regulated by the appropriate interactions between the vesicle (v-) SNARE and the target membrane (t-) SNAREs. We show here that pancreatic acinar cells express the SNAP-25 t-SNARE homolog SNAP-23, and find that this t-SNARE is most highly concentrated on the basolateral plasma membrane while being expressed below detectable levels in endocrine islets within the same tissue. This is the first localization of SNAP-23 within a polarized tissue and suggests that this t-SNAREs may interact with syntaxin-4 to mediate basolateral secretion.


Assuntos
Proteínas de Transporte/análise , Membrana Celular/ultraestrutura , Pâncreas/química , Pâncreas/citologia , Células 3T3 , Animais , Proteínas de Transporte/metabolismo , Fracionamento Celular , Membrana Celular/química , Membrana Celular/metabolismo , Grânulos Citoplasmáticos/química , Grânulos Citoplasmáticos/ultraestrutura , Diabetes Mellitus Experimental/metabolismo , Membranas Intracelulares/química , Membranas Intracelulares/ultraestrutura , Camundongos , Microscopia Confocal , Pâncreas/metabolismo , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Ratos , Ratos Sprague-Dawley
17.
Neurogastroenterol Motil ; 16(3): 325-34, 2004 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15198655

RESUMO

Muscle from the proximal smooth muscle (SM) oesophagus of the cat demonstrates contractions of greater amplitude and greater sensitivity to cholinergic stimulation than muscle from the distal SM oesophagus. In the light of the central role of calcium influx in SM contractility, we hypothesized that regional differences in oesophageal contractility may be associated with differential expression of L-type calcium channels (L(Ca)) along the SM oesophagus. L(Ca) expression was compared between proximal and distal regions of the circular SM oesophagus by Western blots. Patch clamp technique was utilized to study L(Ca) currents. Muscle strip studies assessed L(Ca) contribution to contractile activity. The protein expression of L(Ca) and L(Ca) current density was greater in the proximal than distal region. L(Ca) voltage and time-dependent activation and inactivation curves were similar in cells from both regions. Stimulation of muscle strips with acetylcholine (ACh) in the presence of tetrodotoxin resulted in contractions of greater amplitude in the proximal region. The L(Ca) agonist Bay K 8644 caused a greater increase in ACh-induced contraction amplitude in muscle strips from the proximal region. Therefore, regional myogenic differences in L(Ca) expression along the circular SM oesophageal body exist and may contribute to the nature of oesophageal contractions.


Assuntos
Canais de Cálcio Tipo L/biossíntese , Esôfago/metabolismo , Contração Muscular/fisiologia , Músculo Liso/metabolismo , Peristaltismo/fisiologia , Éster Metílico do Ácido 3-Piridinacarboxílico, 1,4-Di-Hidro-2,6-Dimetil-5-Nitro-4-(2-(Trifluormetil)fenil)/farmacologia , Acetilcolina/farmacologia , Anestésicos Locais/farmacologia , Animais , Western Blotting , Agonistas dos Canais de Cálcio/farmacologia , Canais de Cálcio Tipo L/efeitos dos fármacos , Canais de Cálcio Tipo L/fisiologia , Gatos , Esôfago/efeitos dos fármacos , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Técnicas de Cultura de Órgãos , Técnicas de Patch-Clamp , Peristaltismo/efeitos dos fármacos , Tetrodotoxina/farmacologia
18.
Neurogastroenterol Motil ; 14(4): 383-94, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12213106

RESUMO

Abstract Soluble N-ethylmaleimide-sensitive factors attachment protein receptors (SNAREs), initially found to mediate membrane fusion, have now been shown to also bind and regulate a number of membrane ion channels in neurones and neuroendocrine cells. We recently reported that the SNARE protein SNAP-25 regulates Ca(2+)- activated (K(Ca)) and delays rectifier K(+) channels (K(V)) in oesophageal smooth muscle cells. This raised the possibility that cognate and other SNARE proteins could also be present in the oesophageal smooth muscle cell to regulate these and other functions. Circular muscle tissue sections and single freshly isolated muscle cells from the oesophageal body circular and longitudinal layers, and from lower oesophageal sphincter clasp and sling regions were studied. The subcellular location of SNAP-23, SNAP-25, syntaxins 1 to 4, and vesicle-associated membrane protein (VAMP)-2 were explored using a laser scanning confocal imaging system. Feline oesophageal smooth muscle of all regions examined demonstrated the presence of SNAP-23, SNAP-25, syntaxins 1 to 4, and VAMP-2 on the plasma membrane. The intensity of these syntaxins and SNAP-25/-23 proteins varied between the different muscle groups of the oesophagus. In some regions, some SNARE proteins were also noted in the muscle cell cytoplasm. No differential expression was found for VAMP-2. The differential expression of SNAP-25 and its regulation of K(+) channels indicate the important role of SNAP-25 in regulating the distinct membrane excitability and contractility along the smooth muscle of the oesophagus. This is further contributed by its interactions with the cognate syntaxins, which are also differentially expressed in the muscle groups of the oesophageal body and lower oesophageal sphincter (LOS). These SNARE proteins probably have other functions in the smooth muscle cell, such as regulating vesicular transport processes.


Assuntos
Esôfago/metabolismo , Proteínas de Membrana/biossíntese , Músculo Liso/metabolismo , Proteínas de Transporte Vesicular , Animais , Proteínas de Transporte/metabolismo , Gatos , Esôfago/citologia , Feminino , Masculino , Proteínas de Membrana/metabolismo , Camundongos , Músculo Liso/citologia , Proteínas do Tecido Nervoso/metabolismo , Proteínas Qb-SNARE , Proteínas Qc-SNARE , Proteínas R-SNARE , Proteínas SNARE , Frações Subcelulares/metabolismo , Proteína 25 Associada a Sinaptossoma
19.
Pancreas ; 20(3): 217-26, 2000 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-10766446

RESUMO

The pancreatic acinar cell has been a classic model to study regulated exocytosis occurring at the apical plasma membrane. The acinar cell is also an excellent model with which to study pathologic membrane fusion events, including aberrant zymogen granule fusion with the lysosome and basolateral exocytosis, which are the earliest cellular events of acute pancreatitis. However, despite much effort, little is known about the precise mechanisms that mediate these physiologic and pathologic membrane fusion events until recently. Over the past 5 years, there has been a major advance in the fundamental understanding of vesicle fusion based on the SNARE hypothesis. A basic tenet of the SNARE hypothesis is that the minimal machinery for membrane fusion is a cognate set of v- and t-SNAREs on opposing membranes. A corollary to this hypothesis is that these SNARE proteins are prevented from spontaneous assembly by clamping proteins. Here, the recent developments in the identification of cognate v- and t-SNAREs and clamping proteins are reviewed, which are strategically located to mediate these physiologic exocytic and pathologic fusion events in the pancreatic acinar cell.


Assuntos
Exocitose , Fusão de Membrana , Proteínas de Membrana/fisiologia , Modelos Biológicos , Pâncreas/ultraestrutura , Proteínas de Transporte Vesicular , Animais , Membrana Celular/fisiologia , Humanos , Pancreatite/patologia , Pancreatite/fisiopatologia , Proteínas SNARE
20.
Pancreas ; 9(4): 450-3, 1994 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-7937693

RESUMO

The phenethyl ester analogues of cholecystokinin, OPE and JMV-180, are fully efficacious rat pancreatic secretagogues which, unlike cholecystokinin (CCK), do not elicit supramaximal inhibition of secretion, and stimulate a sustained rise of cytosolic calcium ([Ca2+]i) above basal levels. We have recently shown that low-level protein kinase C (PKC) activation by preincubation of acini with 1 nM 12-O-tetradecanoyl-phorbol-13-acetate (TPA) or minimally secreting concentrations of PKC-activating receptor agonists (1 pM CCK-8, 0.1 microM carbachol or 10 pM bombesin) cause supramaximal inhibition of OPE-stimulated enzyme secretion. We now show that treatment of acini under these conditions also suppresses the sustained rise of [Ca2+]i stimulated by OPE to basal levels in these cells, without changing the initial OPE-stimulated [Ca2+]i peak. The resultant pattern of calcium signalling is similar to that evoked by supramaximal concentrations of native CCK. This suggests that even low concentrations of PKC-activating agonists have the potential to induce inhibitory effects on Ca2+ mobilization and that this kinase is important in generating the supramaximal inhibition observed in response to CCK.


Assuntos
Cálcio/metabolismo , Colecistocinina/análogos & derivados , Pâncreas/metabolismo , Fragmentos de Peptídeos/farmacologia , Sincalida/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Animais , Colecistocinina/farmacologia , Ativação Enzimática , Masculino , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Pâncreas/enzimologia , Proteína Quinase C/agonistas , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA