Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Am J Physiol Endocrinol Metab ; 297(6): E1304-12, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19755672

RESUMO

Fas/Fas ligand belongs to the tumor necrosis factor superfamily of receptors/ligands and is best known for its role in apoptosis. However, recent evidence supports its role in other cellular responses, including proliferation and survival. Although Fas has been implicated as an essential mediator of beta-cell death in the pathogenesis of type 1 diabetes, the essential role of Fas specifically in pancreatic beta-cells has been found to be controversial. Moreover, the role of Fas on beta-cell homeostasis and function is not clear. The objective of this study is to determine the role of Fas specifically in beta-cells under both physiological and diabetes models. Mice with Fas deletion specifically in the beta-cells were generated using the Cre-loxP system. Cre-mediated Fas deletion was under the control of the rat insulin promoter. Absence of Fas in beta-cells leads to complete protection against FasL-induced cell death. However, Fas is not essential in determining beta-cell mass or susceptibility to streptozotocin- or HFD-induced diabetes. Importantly, Fas deletion in beta-cells leads to increased p65 expression, enhanced glucose tolerance, and glucose-stimulated insulin secretion, with increased exocytosis as manifested by increased changes in membrane capacitance and increased expression of Syntaxin1A, VAMP2, and munc18a. Together, our study shows that Fas in the beta-cells indeed plays an essential role in the canonical death receptor-mediated apoptosis but is not essential in regulating beta-cell mass or diabetes development. However, beta-cell Fas is critical in the regulation of glucose homeostasis through regulation of the exocytosis machinery.


Assuntos
Diabetes Mellitus/metabolismo , Proteína Ligante Fas/deficiência , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptor fas/metabolismo , Animais , Apoptose/fisiologia , Proteína Ligante Fas/metabolismo , Feminino , Citometria de Fluxo , Glucose/metabolismo , Teste de Tolerância a Glucose , Secreção de Insulina , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia de Fluorescência , Proteínas Munc18/metabolismo , Técnicas de Patch-Clamp , Sintaxina 1/metabolismo , Proteína 2 Associada à Membrana da Vesícula/metabolismo , eIF-2 Quinase/metabolismo
2.
Mol Cell Biol ; 25(9): 3620-9, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15831467

RESUMO

beta-Cell apoptosis is a key event contributing to the pathogenesis of type 1 diabetes mellitus. In addition to apoptosis being the main mechanism by which beta cells are destroyed, beta-cell apoptosis has been implicated in the initiation of type 1 diabetes mellitus through antigen cross-presentation mechanisms that lead to beta-cell-specific T-cell activation. Caspase-3 is the major effector caspase involved in apoptotic pathways. Despite evidence supporting the importance of beta-cell apoptosis in the pathogenesis of type 1 diabetes, the specific role of caspase-3 in this process is unknown. Here, we show that Caspase-3 knockout (Casp3(-/-) mice were protected from developing diabetes in a multiple-low-dose streptozotocin autoimmune diabetes model. Lymphocyte infiltration of the pancreatic islets was completely absent in Casp3(-/-) mice. To determine the role of caspase-3-dependent apoptosis in disease initiation, a defined antigen-T-cell receptor transgenic system, RIP-GP/P14 double-transgenic mice with Casp3 null mutation, was examined. beta-cell antigen-specific T-cell activation and proliferation were observed only in the pancreatic draining lymph node of RIP-GP/P14/Casp3(+/-) mice, but not in mice lacking caspase-3. Together, our findings demonstrate that caspase-3-mediated beta-cell apoptosis is a requisite step for T-cell priming, a key initiating event in type 1 diabetes.


Assuntos
Apoptose , Caspases/fisiologia , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/imunologia , Ilhotas Pancreáticas/patologia , Animais , Apoptose/genética , Caspase 3 , Caspases/genética , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patologia , Glucagon/análise , Glucagon/metabolismo , Insulina/análise , Insulina/metabolismo , Ilhotas Pancreáticas/enzimologia , Ilhotas Pancreáticas/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Pancrelipase/imunologia , Linfócitos T/imunologia
3.
Diabetes ; 55(2): 435-40, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16443778

RESUMO

Exocytosis of insulin is dependent on the soluble N-ethylmaleimide attachment protein receptor (SNARE) complex proteins in the B-cells. We assessed insulin release as well as gene and protein expression of SNARE complex protein in isolated pancreatic islets of type 2 diabetic patients (n = 4) and nondiabetic control subjects (n = 4). In islets from the diabetic patients, insulin responses to 8.3 and 16.7 mmol/l glucose were markedly reduced compared with control islets (4.7 +/- 0.3 and 8.4 +/- 1.8 vs. 17.5 +/- 0.1 and 24.3 +/- 1.2 microU . islet(-1) . h(-1), respectively; P < 0.001). Western blot analysis revealed decreased amounts of islet SNARE complex and SNARE-modulating proteins in diabetes: syntaxin-1A (21 +/- 5% of control levels), SNAP-25 (12 +/- 4%), VAMP-2 (7 +/- 4%), nSec1 (Munc 18; 34 +/- 13%), Munc 13-1 (27 +/- 4%), and synaptophysin (64 +/- 7%). Microarray gene chip analysis, confirmed by quantitative PCR, showed that gene expression was decreased in diabetes islets: syntaxin-1A (27 +/- 2% of control levels), SNAP-25 (31 +/- 7%), VAMP-2 (18 +/- 3%), nSec1 (27 +/- 5%), synaptotagmin V (24 +/- 2%), and synaptophysin (12 +/- 2%). In conclusion, these data support the view that decreased islet RNA and protein expression of SNARE and SNARE-modulating proteins plays a role in impaired insulin secretion in type 2 diabetic patients. It remains unclear, however, to which extent this defect is primary or secondary to, e.g., glucotoxicity.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas SNARE/biossíntese , Proteínas SNARE/genética , Idoso , Idoso de 80 Anos ou mais , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Glucagon/genética , Glucagon/metabolismo , Humanos , Insulina/genética , Insulina/metabolismo , Masculino , Pessoa de Meia-Idade
4.
Diabetes ; 55(5): 1421-9, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16644700

RESUMO

Munc13-1 is a diacylglycerol (DAG) receptor that is essential for synaptic vesicle priming. We recently showed that Munc13-1 is expressed in rodent and human islet beta-cells and that its levels are reduced in islets of type 2 diabetic humans and rat models, suggesting that Munc13-1 deficiency contributes to the abnormal insulin secretion in diabetes. To unequivocally demonstrate the role of Munc13-1 in insulin secretion, we studied heterozygous Munc13-1 knockout mice (+/-), which exhibited elevated glucose levels during intraperitoneal glucose tolerance tests with corresponding lower serum insulin levels. Munc13-1(+/-) mice exhibited normal insulin tolerance, indicating that a primary islet beta-cell secretory defect is the major cause of their hyperglycemia. Consistently, glucose-stimulated insulin secretion was reduced 50% in isolated Munc13-1(+/-) islets and was only partially rescued by phorbol ester potentiation. The corresponding alterations were minor in mice expressing one allele of a Munc13-1 mutant variant, which does not bind DAG (H567K/+). Capacitance measurements of Munc13-1(+/-) and Munc13-1(H567k/+) islet beta-cells revealed defects in granule priming, including the initial size and refilling of the releasable pools, which become accentuated by phorbol ester potentiation. We conclude that Munc13-1 plays an important role in glucose-stimulated insulin secretion and that Munc13-1 deficiency in the pancreatic islets as occurs in diabetes can reduce insulin secretion sufficient to cause abnormal glucose homeostasis.


Assuntos
Intolerância à Glucose/genética , Insulina/metabolismo , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Cruzamentos Genéticos , Teste de Tolerância a Glucose , Secreção de Insulina , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/fisiologia , Camundongos , Camundongos Knockout , Técnicas de Patch-Clamp , Reação em Cadeia da Polimerase
5.
Endocrinology ; 147(5): 2155-62, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16455778

RESUMO

Glucose regulates pancreatic islet alpha-cell glucagon secretion directly by its metabolism to generate ATP in alpha-cells, and indirectly via stimulation of paracrine release of beta-cell secretory products, particularly insulin. How the cellular substrates of these pathways converge in the alpha-cell is not well known. We recently reported the use of the MIP-GFP (mouse insulin promoter-green fluorescent protein) mouse to reliably identify islet alpha- (non-green cells) and beta-cells (green cells), and characterized their ATP-sensitive K(+) (K(ATP)) channel properties, showing that alpha-cell K(ATP) channels exhibited a 5-fold higher sensitivity to ATP inhibition than beta-cell K(ATP) channels. Here, we show that insulin exerted paracrine regulation of alpha-cells by markedly reducing the sensitivity of alpha-cell K(ATP) channels to ATP (IC(50) = 0.18 and 0.50 mM in absence and presence of insulin, respectively). Insulin also desensitized beta-cell K(ATP) channels to ATP inhibition (IC(50) = 0.84 and 1.23 mM in absence and presence of insulin, respectively). Insulin effects on both islet cell K(ATP) channels were blocked by wortmannin, indicating that insulin acted on the insulin receptor-phosphatidylinositol 3-kinase signaling pathway. Insulin did not affect alpha-cell A-type K(+) currents. Glutamate, known to also inhibit alpha-cell glucagon secretion, did not activate alpha-cell K(ATP) channel opening. We conclude that a major mechanism by which insulin exerts paracrine control on alpha-cells is by modulating its K(ATP) channel sensitivity to ATP block. This may be an underlying basis for the proposed sequential glucose-insulin regulation of alpha-cell glucagon secretion, which becomes distorted in diabetes, leading to dysregulated glucagon secretion.


Assuntos
Trifosfato de Adenosina/química , Trifosfato de Adenosina/metabolismo , Insulina/metabolismo , Potássio/química , Androstadienos/farmacologia , Animais , Peso Corporal , Linhagem Celular , Diabetes Mellitus/metabolismo , Relação Dose-Resposta a Droga , Glucagon/metabolismo , Células Secretoras de Glucagon/citologia , Glucose/metabolismo , Ácido Glutâmico/química , Ácido Glutâmico/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Cobaias , Humanos , Concentração Inibidora 50 , Ilhotas Pancreáticas/metabolismo , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Fosfatidilinositol 3-Quinases/metabolismo , Potássio/metabolismo , Coelhos , Transdução de Sinais , Wortmanina
6.
Diabetes ; 54(9): 2744-54, 2005 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16123365

RESUMO

Soluble N-ethylmaleimide-sensitive factor (NSF) attachment protein receptor (SNARE) protein syntaxin-1A (STX-1A) plays a role not only in exocytosis, but also binds and regulates Ca(2+) and K(+) (voltage-gated K(+) and ATP-sensitive K(+) channels) to influence the sequence of events leading to secretion. Islet levels of STX-1A and cognate SNARE proteins are reduced in type 2 diabetic rodents, suggesting their role in dysregulated insulin secretion contributing to the abnormal glucose homeostasis. We investigated the specific role of STX-1A in pancreatic beta-cells by generating transgenic mice, which express a moderately increased level ( approximately 30% higher) of STX-1A in pancreatic islets (hereafter called STX-1A mice). The STX-1A mice displayed fasting hyperglycemia and a more sustained elevation of plasma glucose levels after an intraperitoneal glucose tolerance test, with correspondingly reduced plasma insulin levels. Surprisingly, beta-cells from the STX-1A male mice also exhibited abnormal insulin tolerance. To unequivocally determine the beta-cell secretory defects, we used single-cell analyses of exocytosis by patch clamp membrane capacitance measurements and ion channel recordings. Depolarization-evoked membrane capacitance increases were reduced in the STX-1A mouse islet beta-cells. The STX-1A mouse also exhibited reduced currents through the Ca(2+) channels but little change in the voltage-gated K(+) channel or ATP-sensitive K(+) channel. These results suggest that fluctuation of islet STX-1A levels in diabetes could influence the pathological and differential regulation of beta-cell ion channels and the exocytotic machinery, collectively contributing to the impaired insulin secretion.


Assuntos
Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatologia , Expressão Gênica/fisiologia , Ilhotas Pancreáticas/metabolismo , Animais , Canais de Cálcio/fisiologia , Modelos Animais de Doenças , Exocitose/fisiologia , Feminino , Teste de Tolerância a Glucose , Insulina/metabolismo , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Canais de Potássio/fisiologia
7.
Biochem J ; 387(Pt 1): 195-202, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15518587

RESUMO

We have shown that SNARE (soluble N-ethylmaleimide-sensitive fusion protein attachment protein receptor) proteins not only participate directly in exocytosis, but also regulate the dominant membrane-repolarizing Kv channels (voltage-gated K+ channels), such as Kv2.1, in pancreatic beta-cells. In a recent report, we demonstrated that WT (wild-type) Syn-1A (syntaxin-1A) inhibits Kv2.1 channel trafficking and gating through binding to the cytoplasmic C-terminus of Kv2.1. During beta-cell exocytosis, Syn-1A converts from a closed form into an open form which reveals its active H3 domain to bind its SNARE partners SNAP-25 (synaptosome-associated protein of 25 kDa) and synaptobrevin. In the present study, we compared the effects of the WT Syn-1A and a mutant open form Syn-1A (L165A, E166A) on Kv2.1 channel trafficking and gating. When co-expressed in HEK-293 cells (human embryonic kidney-293 cells), the open form Syn-1A decreased Kv2.1 current density more than (P<0.05) the WT Syn-1A (166+/-35 and 371+/-93 pA/pF respectively; control=911+/-91 pA/pF). Confocal microscopy and biotinylation experiments showed that both the WT and open form Syn-1A inhibited Kv2.1 expression at the plasma membrane to a similar extent, suggesting that the stronger reduction of Kv2.1 current density by the open form compared with the WT Syn-1A is probably due to a stronger direct inhibition of channel activity. Consistently, dialysis of the recombinant open form Syn-1A protein into Kv2.1-expressing HEK-293 cells caused stronger inhibition of Kv2.1 current amplitude (P<0.05) than the WT Syn-1A protein (73+/-2 and 82+/-3% of the control respectively). We found that the H3 but not H(ABC) domain is the putative active domain of Syn-1A, which bound to and inhibited the Kv2.1 channel. When co-expressed in HEK-293 cells, the open-form Syn-1A slowed down Kv2.1 channel activation (tau=12.3+/-0.8 ms) much more than (P<0.05) WT Syn-1A (tau=7.9+/-0.8 ms; control tau=5.5+/-0.6 ms). In addition, only the open form Syn-1A, but not the WT Syn-1A, caused a significant (P<0.05) left-shift in the steady-state inactivation curve (V(1/2)=33.1+/-1.3 and -29.4+/-1.1 mV respectively; control V(1/2)=-24.8+/-2 mV). The present study therefore indicates that the open form of Syn-1A is more potent than the WT Syn-1A in inhibiting the Kv2.1 channel. Such stronger inhibition by the open form of Syn-1A may limit K+ efflux and thus decelerate membrane repolarization during exocytosis, leading to optimization of insulin release.


Assuntos
Antígenos de Superfície/química , Antígenos de Superfície/fisiologia , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/metabolismo , Antígenos de Superfície/biossíntese , Canais de Potássio de Retificação Tardia , Humanos , Ativação do Canal Iônico/fisiologia , Rim/química , Rim/citologia , Rim/embriologia , Rim/metabolismo , Técnicas de Patch-Clamp/métodos , Canais de Potássio de Abertura Dependente da Tensão da Membrana/biossíntese , Canais de Potássio de Abertura Dependente da Tensão da Membrana/genética , Estrutura Quaternária de Proteína/fisiologia , Canais de Potássio Shab , Sintaxina 1 , Transfecção/métodos
8.
Endocrinology ; 146(11): 4766-75, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16109783

RESUMO

We recently reported a transgenic [mouse insulin promoter (MIP)-green fluorescent protein (GFP)] mouse in which GFP expression is targeted to the pancreatic islet beta-cells to enable convenient identification of beta-cells as green cells. The GFP-expressing beta-cells of the MIP-GFP mouse were functionally indistinguishable from beta-cells of normal mice. Here we characterized the ionic channel properties and exocytosis of MIP-GFP mouse islet beta- and alpha-cells. Beta-cells displayed delayed rectifying K+ and high-voltage-activated Ca2+ channels and exhibited Na+ currents only at hyperpolarized holding potential. Alpha-cells were nongreen and had both A-type and delayed rectifier K+ channels, both low-voltage-activated and high-voltage-activated Ca2+ channels, and displayed Na+ currents readily at -70 mV holding potential. Alpha-cells had ATP-sensitive K+ channel (KATP) channel density as high as that in beta-cells, and, surprisingly, alpha-cell KATP channels were more sensitive to ATP inhibition (IC50=0.16+/-0.03 mM) than beta-cell KATP channels (IC50=0.86+/-0.10 mM). Whereas alpha-cells were rather uniform in size [2-4.5 picofarad (pF)], beta-cells varied vastly in size (2-12 pF). Of note, small beta-cells (<4.5 pF) showed little exocytosis, whereas medium beta-cells (5-8 pF) exhibited vigorous exocytosis, but large beta-cells (>8 pF) had weaker exocytosis. We found no correlation between beta-cell size and their Ca2+ channel density, suggesting that Ca2+ influx may not be the cause of the heterogeneity in exocytotic responses. The MIP-GFP mouse therefore offers potential to further explore the functional heterogeneity in beta-cells of different sizes. The MIP-GFP mouse islet is therefore a reliable model to efficiently examine alpha-cell and beta-cell physiology and should greatly facilitate examination of their pathophysiology when the MIP-GFP mice are crossed with diabetic models.


Assuntos
Proteínas de Fluorescência Verde/genética , Insulina/genética , Ilhotas Pancreáticas/fisiologia , Camundongos Transgênicos/fisiologia , Regiões Promotoras Genéticas , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Canais de Cálcio/fisiologia , Eletrofisiologia , Exocitose , Técnicas In Vitro , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Microscopia Confocal , Microscopia de Fluorescência , Canais de Potássio/efeitos dos fármacos , Canais de Potássio/fisiologia , Canais de Potássio de Abertura Dependente da Tensão da Membrana/fisiologia , Canais de Sódio/fisiologia
9.
Diabetes ; 51(5): 1425-36, 2002 May.
Artigo em Inglês | MEDLINE | ID: mdl-11978639

RESUMO

Cognate soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins are now known to associate the secretory vesicle with both the target plasma membrane and Ca(2+) channels in order to mediate the sequence of events leading to exocytosis in neurons and neuroendocrine cells. Neuroendocrine cells, particularly insulin-secreting islet beta-cells, t-SNARE proteins, 25-kDa synaptosomal-associated protein (SNAP-25), and syntaxin 1A, independently inhibit the L-type Ca(2+) channel (L(Ca)). However, when both are present, they actually exhibit stimulatory actions on the L(Ca). This suggests that the positive regulation of the L(Ca) is conferred by a multi-SNARE protein complex. We hypothesized an alternate explanation, which is that each of these SNARE proteins possess distinct inhibitory and stimulatory domains that act on the L(Ca). These SNARE proteins were recently shown to bind the Lc(753-893) domain corresponding to the II and III intracellular loop of the alpha1C subunit of the L(Ca). In this study, using patch-clamp methods on primary pancreatic beta-cells and insulinoma HIT-T15 cells, we examined the functional interactions of the botulinum neurotoxin A (BoNT/A) cleavage products of SNAP-25, including NH(2)-terminal (1-197 amino acids) and COOH-terminal (amino acid 198-206) domains, on the L(Ca), particularly at the Lc(753-893) domain. Intracellular application of SNAP-25(1-206) in primary beta-cells decreased L(Ca) currents by approximately 15%. The reduction in L(Ca) currents was counteracted by coapplication of Lc(753-893). Overexpression or injection of wild-type SNAP-25 in HIT cells reduced L(Ca) currents by approximately 30%, and this inhibition was also blocked by the recombinant Lc(753-893) peptide. Expression of BoNT/A surprisingly caused an even greater reduction of L(Ca) currents (by 41%), suggesting that the BoNT/A cleavage products of SNAP-25 might possess distinct inhibitory and positive regulatory domains. Indeed, expression of SNAP-25(1-197) increased L(Ca) currents (by 19% at 10 mV), and these effects were blocked by the Lc(753-893) peptide. In contrast, injection of SNAP-25(198-206) peptide into untransfected cells inhibited L(Ca) currents (by 47%), and more remarkably, these inhibitory effects dominated over the stimulatory effects of SNAP-25(1-197) overexpression (by 34%). Therefore, the SNARE protein SNAP-25 possesses distinct inhibitory and stimulatory domains that act on the L(Ca). The COOH-terminal 197-206 domain of SNAP-25, whose inhibitory actions dominate over the opposing stimulatory NH(2)-terminal domain, likely confers the inhibitory actions of SNAP-25 on the L(Ca). We postulate that the eventual accelerated proteolysis of SNAP-25 brought about by BoNT/A cleavage allows the relatively intact NH(2)-terminal SNAP-25 domain to assert its stimulatory action on the L(Ca) to increase Ca(2+) influx, and this could in part explain the observed weak or inconsistent inhibitory effects of BoNT/A on insulin secretion. The present study suggests that distinct domains within SNAP-25 modulate L(C) subtype Ca(2+) channel activity in both primary beta-cells and insulinoma HIT-T15 cells.


Assuntos
Canais de Cálcio Tipo L/metabolismo , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/genética , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Animais , Toxinas Botulínicas Tipo A , Cálcio/metabolismo , Linhagem Celular , Proteínas de Fluorescência Verde , Indicadores e Reagentes/metabolismo , Ativação do Canal Iônico/efeitos dos fármacos , Ativação do Canal Iônico/fisiologia , Ilhotas Pancreáticas/citologia , Proteínas Luminescentes/genética , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Proteínas de Membrana/farmacologia , Proteínas do Tecido Nervoso/farmacologia , Fármacos Neuromusculares , Técnicas de Patch-Clamp , Fragmentos de Peptídeos/metabolismo , Fragmentos de Peptídeos/farmacologia , Estrutura Terciária de Proteína , Proteína 25 Associada a Sinaptossoma , Transfecção
10.
Endocrinology ; 143(11): 4218-26, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12399415

RESUMO

The role of glucotoxicity in dysregulation of islet exocytotic soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) complex proteins and insulin response was explored in the hyperglycemic Goto-Kakizaki (GK) rat. Syntaxin-1A and vesicle-associated membrane protein isoform 2, which drive insulin granule exocytotic fusion, and the associated nSec1, which modulates the SNARE complex assembly, were diminished in GK pancreatic islets to approximately 40% of the levels in control Wistar rat islets. Phlorizin treatment (12 d) induced normoglycemic control in GK rats, resulting in partial restoration of the insulin response to glucose. Furthermore, islet SNARE complex and nSec1 proteins increased by about 40%. Phlorizin treatment did not affect levels of islet SNARE proteins in controls or on the same SNARE complex proteins in GK rat brain. To examine the role of hyperglycemia per se, GK and control rat islets were exposed for 5 d in culture to 5.5 and 16.7 mM glucose. High glucose treatment greatly increased the levels of synaptosomal-associated membrane protein of 25 kDa and, less markedly, the levels of syntaxin-1A and nSec1 in control islets more than in GK rat islets, whereas levels were reduced in both. This was accompanied by sustained impairment of the insulin response to glucose in GK islets and a normal response in control islets. Thus, GK islets demonstrate dysregulation of SNARE protein expression, and their compensatory increase by high glucose exposure is abrogated. Conversely, normoglycemic control results in partial replenishment of these critical components of the insulin exocytotic machinery and improvement in the insulin response. We propose that dysregulation of SNARE proteins is an important mechanism behind glucotoxicity-mediated impairment of the insulin response to glucose.


Assuntos
Proteínas de Transporte/genética , Expressão Gênica/efeitos dos fármacos , Glucose/farmacologia , Ilhotas Pancreáticas/metabolismo , Proteínas de Membrana/genética , Florizina/farmacologia , Proteínas de Transporte Vesicular , Animais , Proteínas de Transporte/análise , Técnicas de Cultura , Hiperglicemia/metabolismo , Immunoblotting , Insulina/metabolismo , Secreção de Insulina , Ilhotas Pancreáticas/química , Masculino , Proteínas de Membrana/análise , Proteínas Sensíveis a N-Etilmaleimida , Ratos , Ratos Wistar , Proteínas de Ligação a Fator Solúvel Sensível a N-Etilmaleimida
11.
Am J Physiol Endocrinol Metab ; 296(6): E1251-61, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19223654

RESUMO

Thioredoxin-interacting protein (TxNIP) is an endogenous inhibitor of thioredoxin, a ubiquitous thiol oxidoreductase, that regulates cellular redox status. Diabetic mice exhibit increased expression of TxNIP in pancreatic islets, and recent studies suggest that TxNIP is a proapoptotic factor in beta-cells that may contribute to the development of diabetes. Here, we examined the role of TxNIP deficiency in vivo in the development of insulin-deficient diabetes and whether it impacted on pancreatic beta-cell mass and/or insulin secretion. TxNIP-deficient (Hcb-19/TxNIP(-/-)) mice had lower baseline glycemia, higher circulating insulin concentrations, and higher total pancreatic insulin content and beta-cell mass than control mice (C3H). Hcb-19/TxNIP(-/-) did not develop hyperglycemia when injected with standard multiple low doses of streptozotocin (STZ), in contrast to C3H controls. Surprisingly, although beta-cell mass remained higher in Hcb-19/TxNIP(-/-) mice compared with C3H after STZ exposure, the relative decrease induced by STZ was as great or even greater in the TxNIP-deficient animals. Consistently, cultured pancreatic INS-1 cells transfected with small-interfering RNA against TxNIP were more sensitive to cell death induced by direct exposure to STZ or to the combination of inflammatory cytokines interleukin-1beta, interferon-gamma, and tumor necrosis factor-alpha. Furthermore, when corrected for insulin content, isolated pancreatic islets from TxNIP(-/-) mice exhibited reduced glucose-induced insulin secretion. These data indicate that TxNIP functions as a regulator of beta-cell mass and influences insulin secretion. In conclusion, the relative resistance of TxNIP-deficient mice to STZ-induced diabetes appears to be because of an increase in beta-cell mass. However, TxNIP deficiency is associated with sensitization to STZ- and cytokine-induced beta-cell death, indicating complex regulatory roles of TxNIP under different physiological and pathological conditions.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/patologia , Células Secretoras de Insulina/citologia , Tiorredoxinas/genética , Tiorredoxinas/metabolismo , Animais , Antibióticos Antineoplásicos/farmacologia , Apoptose/fisiologia , Proteínas de Ciclo Celular , Linhagem Celular , Citocinas/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Diabetes Mellitus Tipo 1/fisiopatologia , Relação Dose-Resposta a Droga , Glucose/farmacologia , Marcação In Situ das Extremidades Cortadas , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C3H , Camundongos Mutantes , Ratos , Estreptozocina/farmacologia
12.
Pancreas ; 34(3): 359-63, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17414060

RESUMO

OBJECTIVES: We examined how lipotoxicity contributes to pancreatic beta-cell secretory dysfunction. METHODS: Effects of palmitate (0.2 mmol/L) were assessed on insulin secretion and soluble N-ethylmaleimide-sensitive factor attachment protein receptor exocytotic machinery in isolated pancreatic islets of type 2 diabetic Goto-Kakizaki (GK) rats and control Wistar (W) rats. RESULTS: One-day palmitate treatment enhanced basal glucose (3.3 mmol/L)-mediated insulin release 5-fold in W and 3.3-fold in GK islets, but had no effect at high glucose (16.7 mmol/L) on W islets while enhancing GK islet insulin release 2-fold. After 3-day palmitate treatment, high-glucose-induced insulin release in W islets was reduced (by 69%), whereas in GK islets, it increased 2-fold. Insulin response to arginine was reduced in both islet types, but more so in GK islets. Exocytotic proteins (syntaxin 1A, VAMP-2, SNAP-25, nSec1) were reduced in GK islets by 56% to 69% compared with W islets. In W islets, palmitate treatment caused no changes in the levels of these proteins but increased actin levels. In GK islets, whereas 1-day palmitate treatment had no effect, 3-day treatment further reduced SNAP-25 and nSec1 levels. CONCLUSIONS: Lipotoxic-induced secretory insufficiency in normal islets may be attributed to lack of compensatory increase in levels of exocytotic proteins and/or excess actin. However, in GK islets, palmitate treatment moderately enhanced insulin secretion, likely by acting on proximal metabolic pathways capable of compensating for the defective soluble N-ethylmaleimide-sensitive factor attachment protein receptor exocytotic machinery. These results were different from prolonged glucose treatment we previously reported, indicating differences between glucotoxic and lipotoxic actions on the insulin secretory machinery.


Assuntos
Exocitose/efeitos dos fármacos , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Ácido Palmítico/farmacologia , Proteínas/metabolismo , Animais , Glucose/farmacologia , Secreção de Insulina , Ilhotas Pancreáticas/efeitos dos fármacos , Ratos , Ratos Mutantes
13.
Diabetes ; 56(10): 2579-88, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17639022

RESUMO

OBJECTIVE: Glucagon-like peptide-1 (GLP-1) rescues insulin secretory deficiency in type 2 diabetes partly via cAMP actions on exchange protein directly activated by cAMP (Epac2) and protein kinase A (PKA)-activated Rab3A-interacting molecule 2 (Rim2). We had reported that haplodeficient Munc13-1(+/-) mouse islet beta-cells exhibited reduced insulin secretion, causing glucose intolerance. Munc13-1 binds Epac2 and Rim2, but their functional interactions remain unclear. RESEARCH DESIGN AND METHODS: We used Munc13-1(+/-) islet beta-cells to examine the functional interactions between Munc13-1 and Epac2 and PKA. GLP-1 stimulation of Munc13-1(+/-) islets normalized the reduced biphasic insulin secretion by its actions on intact islet cAMP production and normal Epac2 and Rim2 levels. RESULTS: To determine which exocytotic steps caused by Munc13-1 deficiency are rescued by Epac2 and PKA, we used patch-clamp capacitance measurements, showing that 1) cAMP restored the reduced readily releasable pool (RRP) and partially restored refilling of a releasable pool of vesicles in Munc13-1(+/-) beta-cells, 2) Epac-selective agonist [8-(4-chloro-phenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate] partially restored the reduced RRP and refilling of a releasable pool of vesicles, and 3) PKA blockade by H89 (leaving Epac intact) impaired cAMP ability to restore the RRP and refilling of a releasable pool of vesicles. Conversely, PKA-selective agonist (N(6)-benzoyladenosine-cAMP) completely restored RRP and partially restored refilling of a releasable pool of vesicles. To determine specific contributions within Epac-Rim2-Munc13-1 interaction sites accounting for cAMP rescue of exocytosis caused by Munc13-1 deficiency, we found that blockade of Rim2-Munc13-1 interaction with Rim-Munc13-1-binding domain peptide abolished cAMP rescue, whereas blockade of Epac-Rim2 interaction with Rim2-PDZ peptide only moderately reduced refilling with little effect on RRP. CONCLUSIONS: cAMP rescue of priming defects caused by Munc13-1 deficiency via Epac and PKA signaling pathways requires downstream Munc13-1-Rim2 interaction.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Exocitose/fisiologia , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Células Secretoras de Insulina/fisiologia , Insulina/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Animais , AMP Cíclico/metabolismo , Exocitose/efeitos dos fármacos , Insulina/deficiência , Secreção de Insulina , Células Secretoras de Insulina/efeitos dos fármacos , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Proteínas do Tecido Nervoso/genética , Técnicas de Patch-Clamp
14.
Diabetes ; 56(9): 2302-11, 2007 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-17563067

RESUMO

Inadequate pancreatic beta-cell mass resulting from excessive beta-cell apoptosis is a key defect in type 1 and type 2 diabetes. Caspases are the major molecules involved in apoptosis; however, in vivo roles of specific caspases in diabetes are unclear. The purpose of this study is to examine the role of Caspase (Casp)8 in beta-cells in vivo. Using the Cre-loxP system, mice lacking Casp8 in beta-cells (RIPcre(+)Casp8(fl/fl) mice) were generated to address the role of Casp8 in beta-cells in physiological and diabetes models. We show that islets isolated from RIPcre(+)Casp8(fl/fl) mice were protected from Fas ligand (FasL)-and ceramide-induced cell death. Furthermore, RIPcre(+)Casp8(fl/fl) mice were protected from in vivo models of type 1 and type 2 diabetes. In addition to being the central mediator of apoptosis in diabetes models, we show that Casp8 is critical for maintenance of beta-cell mass under physiological conditions. With aging, RIPcre(+)Casp8(fl/fl) mice gradually develop hyperglycemia and a concomitant decline in beta-cell mass. Their islets display decreased expression of molecules involved in insulin/IGF-I signaling and show decreased pancreatic duodenal homeobox-1 and cAMP response element binding protein expression. At the level of individual islets, we observed increased insulin secretory capacity associated with increased expression of exocytotic proteins. Our results show distinct context-specific roles of Casp8 in physiological and disease states; Casp8 is essential for beta-cell apoptosis in type 1 and type 2 diabetes models and in regulating beta-cell mass and insulin secretion under physiological conditions.


Assuntos
Caspase 8/genética , Caspase 8/metabolismo , Diabetes Mellitus Experimental/enzimologia , Células Secretoras de Insulina/enzimologia , Animais , Glicemia/metabolismo , Morte Celular , Separação Celular , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 1/enzimologia , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 2/enzimologia , Diabetes Mellitus Tipo 2/patologia , Células Secretoras de Insulina/patologia , Camundongos , Camundongos Endogâmicos C57BL , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mol Pharmacol ; 70(3): 818-28, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16754785

RESUMO

We have hypothesized that the plasma membrane protein components of the exocytotic soluble N-ethylmaleimide-sensitive factor attachment protein (SNAP) receptor (SNARE) complex, syntaxin 1A and SNAP-25, distinctly regulate different voltage-gated K+ (Kv) channels that are differentially distributed. Neuroendocrine islet cells (alpha, beta, delta) uniformly contain both syntaxin 1A and SNAP-25. However, using immunohistochemistry, we show that the different pancreatic islet cells contain distinct dominant Kv channels, including Kv2.1 in beta cells and Kv2.2 in alpha and delta cells, whose interactions with the SNARE proteins would, respectively regulate insulin, glucagon and somatostatin secretion. We therefore examined the regulation by syntaxin 1A and SNAP-25 of these two channels. We have shown that Kv2.1 interacts with syntaxin 1A and SNAP-25 and, based on studies in oocytes, suggested a model of two distinct modes of interaction of syntaxin 1A and the complex syntaxin 1A/SNAP-25 with the C terminus of the channel. Here, we characterized the interactions of syntaxin 1A and SNAP-25 with Kv2.2 which is highly homologous to Kv2.1, except for the C-terminus. Comparative two-electrode voltage clamp analysis in oocytes between Kv2.2 and Kv2.1 shows that Kv2.2 interacts only with syntaxin 1A and, in contrast to Kv2.1, it does not interact with the syntaxin 1A/SNAP-25 complex and hence is not sensitive to the assembly/disassembly state of the complex. The distinct regulation of these closely related channels by SNAREs may be attributed to differences in their C termini. Together with the differential distribution of these channels among islet cells, their distinct regulation suggests that the documented profound down-regulation of islet SNARE levels in diabetes could distort islet cell ion channels and secretory responses in different ways, ultimately contributing to the abnormal glucose homeostasis.


Assuntos
Ativação do Canal Iônico/fisiologia , Ilhotas Pancreáticas/metabolismo , Canais de Potássio Shab/metabolismo , Proteína 25 Associada a Sinaptossoma/metabolismo , Sintaxina 1/metabolismo , Animais , Humanos , Cinética , Oócitos , Células PC12 , Ligação Proteica , Ratos , Solubilidade , Xenopus laevis
16.
Biochem Biophys Res Commun ; 345(1): 340-4, 2006 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-16678791

RESUMO

In low or absence of glucose, alpha-cells generate rhythmic action potentials and secrete glucagon. alpha-Cell T-type Ca(2+) channels are believed to be pacemaker channels, which are expected to open near the resting membrane potential (around -60 mV) to initiate a small depolarization. A previous publication, however, showed that alpha-cell T-type Ca(2+) channels have an activation threshold of -40 mV, which does not appear to fulfill their role as pacemakers. In this work, we investigated the Ca(2+) channel characteristics in alpha-cells of mouse-insulin-promoter green-fluorescent-protein (MIP-GFP) mouse. The beta-cells of MIP-GFP were conveniently distinguished as green cells, while immunostaining indicated that the majority of non-green cells were alpha-cells. We found that majority of alpha-cells possessed T-type Ca(2+) channels having an activation threshold of -40 mV; these cells also had high-voltage-activated (HVA) Ca(2+) channels (activation threshold of -20 mV). A novel finding here is that a minority of alpha-cells had T-type Ca(2+) channels with an activation threshold of -60 mV. This minor population of alpha-cells was, surprisingly, devoid of HVA Ca(2+) channels. We suggest that this alpha-cell subpopulation may act as pacemaker cells in low or absence of glucose.


Assuntos
Canais de Cálcio/fisiologia , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Células Secretoras de Glucagon/classificação , Células Secretoras de Glucagon/fisiologia , Ativação do Canal Iônico/fisiologia , Potenciais da Membrana/fisiologia , Potenciais de Ação/fisiologia , Animais , Relógios Biológicos/fisiologia , Células Cultivadas , Células Secretoras de Glucagon/citologia , Camundongos
17.
Am J Physiol Gastrointest Liver Physiol ; 288(6): G1233-40, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15691872

RESUMO

Potassium channels are important contributors to membrane excitability in smooth muscles. There are regional differences in resting membrane potential and K(+)-channel density along the length of the feline circular smooth muscle esophagus. The aim of this study was to assess responses of K(+)-channel currents to cholinergic (ACh) stimulation along the length of the feline circular smooth muscle esophageal body. Perforated patch-clamp technique assessed K(+)-channel responses to ACh stimulation in isolated smooth muscle cells from the circular muscle layer of the esophageal body at 2 (distal)- and 4-cm (proximal) sites above the lower esophageal sphincter. Western immunoblots assessed ion channel and receptor expression. ACh stimulation produced a transient increase in outward current followed by inhibition of spontaneous transient outward currents. These ACh-induced currents were abolished by blockers of large-conductance Ca(2+)-dependent K(+) channels (BK(Ca)). Distal cells demonstrated a greater peak current density in outward current than cells from the proximal region and a longer-lasting outward current increase. These responses were abolished by atropine and the specific M(3) receptor antagonist 4-DAMP but not the M(1) receptor antagonist pirenzipine or the M(2) receptor antagonist methoctramine. BK(Ca) expression along the smooth muscle esophagus was similar, but M(3) receptor expression was greater in the distal region. Therefore, ACh can differentially activate a potassium channel (BK(Ca)) current along the smooth muscle esophagus. This activation probably occurs through release of intracellular calcium via an M(3) pathway and has the potential to modulate the timing and amplitude of peristaltic contraction along the esophagus.


Assuntos
Acetilcolina/farmacologia , Esôfago/fisiologia , Músculo Liso/fisiologia , Canais de Potássio/fisiologia , Receptor Muscarínico M3/fisiologia , Animais , Gatos , Eletrofisiologia , Esôfago/anatomia & histologia , Potenciais da Membrana , Antagonistas Muscarínicos/farmacologia , Piperidinas/farmacologia , Pirenzepina/farmacologia
18.
Pancreas ; 28(4): 395-400, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15097857

RESUMO

The minimal machinery for fusion of secretory vesicles with the cell membrane is a cognate set of v- and t-SNAREs on opposing membranes. Spontaneous SNARE complex assembly leading to unregulated membrane fusion is prevented by Munc18 proteins that bind membrane SNAREs syntaxins. Munc18 blocks syntaxin interactions with cognate SNARE proteins and thereby act as an inhibitor of exocytosis. The pancreatic acinar cell contains several sets of cognate SNAREs and Munc18 proteins that mediate the distinct exocytic events. We had reported that in the rat pancreas, Munc18c co-localizes with t-SNAREs syntaxin4 and SNAP23 on the acinar cell basolateral plasma membrane. Under conditions that induce pancreatitis in vivo, displacement of Munc18c from the basolateral plasma membrane relieved its blockade of SNARE-mediated membrane fusion in this region and thereby redirected apical exocytosis to the basal membrane surface. Here we show in a case of human mild alcoholic chronic pancreatitis that Munc18c is also displaced from the plasma membrane of intact acinar cells, which would render these cells receptive to pathologic basolateral exocytosis and further episodes of pancreatitis.


Assuntos
Membrana Celular/química , Proteínas do Tecido Nervoso , Pâncreas/química , Pancreatite Alcoólica/metabolismo , Proteínas/análise , Proteínas de Transporte Vesicular , Proteínas de Transporte/análise , Polaridade Celular , Humanos , Proteínas de Membrana/análise , Proteínas Munc18 , Pâncreas/citologia , Pâncreas/patologia , Pancreatite Alcoólica/patologia , Proteínas Qa-SNARE , Proteínas Qb-SNARE , Proteínas Qc-SNARE
19.
Pancreas ; 25(4): e56-63, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12409842

RESUMO

INTRODUCTION AND AIM: We used the amphicrine AR42J as an excellent model to study the differentiation of the secretory machinery of pancreatic endocrine and exocrine cells. Dexamethasone treatment induced the AR42J to differentiate towards the exocrine phenotype capable of secreting amylase in response to cholecystokinin. In contrast, activin A plus hepatocyte growth factor treatment of a subclone of AR42J, AR42J-B13, induced this cell to differentiate morphologically and functionally toward an insulin-containing and insulin-secreting endocrine phenotype. We took advantage of these unique properties of selective exocrine and endocrine induction of the AR42J to reveal which distinct combinations of exocytic SNARE complex proteins (syntaxin, SNAP-25 and VAMP) and associated Munc18 proteins were preferentially expressed to play a role in enzyme and insulin secretion. RESULTS AND CONCLUSION: To our surprise, both endocrine and exocrine induction of AR42J and AR42J-B13 caused very similar upregulation in the expression of the exocytic member isoforms of the syntaxin, SNAP-25, VAMP, and Munc18 families. We conclude that whereas the differentiation of the proximal components of the secretory machinery of the exocrine acinar and endocrine islet beta-cells is distinct, the differentiation of the distal components of exocytosis between these two cell types is very similar.


Assuntos
Proteínas de Membrana/biossíntese , Pâncreas/metabolismo , Biossíntese de Proteínas , Proteínas de Transporte Vesicular , Ativinas/farmacologia , Amilases/metabolismo , Diferenciação Celular , Linhagem Celular , Dexametasona/farmacologia , Fator de Crescimento de Hepatócito/farmacologia , Subunidades beta de Inibinas/farmacologia , Insulina/metabolismo , Proteínas Munc18 , Proteínas do Tecido Nervoso/biossíntese , Pâncreas/citologia , Pâncreas/efeitos dos fármacos , Fenótipo , Proteínas Qa-SNARE , Proteínas R-SNARE , Proteína 25 Associada a Sinaptossoma , Regulação para Cima
20.
Biochem Biophys Res Commun ; 292(4): 980-6, 2002 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-11944911

RESUMO

The examination of insulin exocytosis at the single cell level by conventional electrophysiologic and amperometric methods possesses inherent limitations, and may not accurately reflect the morphologic events of exocytosis of the insulin granule. To overcome some of these limitations, we show by epifluorescent microscopy of a fluorescent dye, FM1-43, its incorporation into the plasma membrane and oncoming insulin granules undergoing exocytosis, and their core proteins. Using this method, we tracked exocytosis in real-time in insulinoma INS-1 and single rat islet beta cells in response to KCl and glucose. We observed both single transient and multi-stepwise increases in membrane FM1-43 fluorescence, suggesting single granule exocytosis as well as sequential and compound exocytosis, respectively. Confocal microscopy of nonpermeabilized cells shows that some of the exocytosed insulin granules labeled by the FM1-43 dye could also be labeled with insulin antibodies, suggesting prolonged openings of the fusion pores and slow dissolution of the granule core proteins on the membrane surface.


Assuntos
Exocitose/fisiologia , Ilhotas Pancreáticas/metabolismo , Animais , Membrana Celular/efeitos dos fármacos , Células Cultivadas , Exocitose/efeitos dos fármacos , Corantes Fluorescentes , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina , Insulinoma/tratamento farmacológico , Insulinoma/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Cloreto de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Vesículas Secretórias/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa