Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Mol Metab ; 6(10): 1161-1172, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-29031717

RESUMO

OBJECTIVE: Hyperglucagonemia is present in many forms of diabetes and contributes to hyperglycemia, and glucagon suppression can ameliorate diabetes in mice. Leptin, a glucagon suppressor, can also reverse diabetes in rodents. Lipid nanoparticle (LNP) delivery of small interfering RNA (siRNA) effectively targets the liver and is in clinical trials for the treatment of various diseases. We compared the effectiveness of glucagon receptor (Gcgr)-siRNA delivered via LNPs to leptin in two mouse models of diabetes. METHODS: Gcgr siRNA encapsulated into LNPs or leptin was administered to mice with diabetes due to injection of the ß-cell toxin streptozotocin (STZ) alone or combined with high fat diet (HFD/STZ). RESULTS: In STZ-diabetic mice, a single injection of Gcgr siRNA lowered blood glucose levels for 3 weeks, improved glucose tolerance, and normalized plasma ketones levels, while leptin therapy normalized blood glucose levels, oral glucose tolerance, and plasma ketones, and suppressed lipid metabolism. In contrast, in HFD/STZ-diabetic mice, Gcgr siRNA lowered blood glucose levels for 2 months, improved oral glucose tolerance, and reduced HbA1c, while leptin had no beneficial effects. CONCLUSIONS: While leptin may be more effective than Gcgr siRNA at normalizing both glucose and lipid metabolism in STZ diabetes, Gcgr siRNA is more effective at reducing blood glucose levels in HFD/STZ diabetes.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Nanopartículas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , Receptores de Glucagon/genética , Animais , Diabetes Mellitus Experimental/terapia , Dieta Hiperlipídica , Glucagon/sangue , Homeostase , Hiperglicemia/sangue , Hiperglicemia/genética , Insulina/sangue , Leptina/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nanopartículas/química , RNA Interferente Pequeno/genética , Receptores de Glucagon/antagonistas & inibidores , Receptores de Glucagon/biossíntese
2.
Mod Pathol ; 28(3): 391-402, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25216224

RESUMO

Glucagon-like-peptide-1 (GLP1) analogs may induce thyroid or pancreatic diseases in animals, raising questions about their use in diabetic patients. There is, however, controversy regarding expression of GLP1 receptors (GLP1R) in human normal and diseased thyroid and pancreas. Here, 221 human thyroid and pancreas samples were analyzed for GLP1R immunohistochemistry and compared with quantitative in vitro GLP1R autoradiography. Neither normal nor hyperplastic human thyroids containing parafollicular C cells express GLP1R with either method. Papillary thyroid cancer do not, and medullary thyroid carcinomas rarely express GLP1R. Insulin- and somatostatin-producing cells in the normal pancreas express a high density of GLP1R, whereas acinar cells express them in low amounts. Ductal epithelial cells do not express GLP1R. All benign insulinomas express high densities of GLP1R, whereas malignant insulinomas rarely express them. All ductal pancreatic carcinomas are GLP1R negative, whereas 6/20 PanIN 1/2 and 0/12 PanIN 3 express GLP1R. Therefore, normal thyroid, including normal and hyperplastic C cells, or papillary thyroid cancer are not targets for GLP1 analogs in humans. Conversely, all pancreatic insulin- and somatostatin-producing cells are physiological GLP1 targets, as well as most acini. As normal ductal epithelial cells or PanIN 3 or ductal pancreatic carcinomas do not express GLP1R, it seems unlikely that GLP1R is related to neoplastic transformation in pancreas. GLP1R-positive medullary thyroid carcinomas and all benign insulinomas are candidates for in vivo GLP1R targeting.


Assuntos
Pâncreas/metabolismo , Receptores de Glucagon/biossíntese , Glândula Tireoide/metabolismo , Autorradiografia , Western Blotting , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Imuno-Histoquímica , Neoplasias Pancreáticas/metabolismo , Doenças da Glândula Tireoide/metabolismo , Neoplasias da Glândula Tireoide/metabolismo
3.
J Biol Chem ; 290(2): 1233-43, 2015 Jan 09.
Artigo em Inglês | MEDLINE | ID: mdl-25451942

RESUMO

The signaling capacity of seven-transmembrane/G-protein-coupled receptors (7TM/GPCRs) can be regulated through ligand-mediated receptor trafficking. Classically, the recycling of internalized receptors is associated with resensitization, whereas receptor degradation terminates signaling. We have shown previously that the incretin glucagon-like peptide-1 receptor (GLP-1R) internalizes fast and is primarily resensitized through recycling back to the cell surface. GLP-1R is expressed in pancreatic islets together with the closely related glucose-dependent insulinotropic polypeptide (GIPR) and glucagon (GCGR) receptors. The interaction and cross-talk between coexpressed receptors is a wide phenomenon of the 7TM/GPCR superfamily. Numerous reports show functional consequences for signaling and trafficking of the involved receptors. On the basis of the high structural similarity and tissue coexpression, we here investigated the potential cross-talk between GLP-1R and GIPR or GCGR in both trafficking and signaling pathways. Using a real-time time-resolved FRET-based internalization assay, we show that GLP-1R, GIPR, and GCGR internalize with differential properties. Remarkably, upon coexpression of the internalizing GLP-1R and the non-internalizing GIPR, GLP-1-mediated GLP-1R internalization was impaired in a GIPR concentration-dependent manner. As a functional consequence of such impaired internalization capability, GLP-1-mediated GLP-1R signaling was abrogated. A similar compromised signaling was found when GLP-1R internalization was abrogated by a dominant-negative version of dynamin (dynamin-1 K44E), which provides a mechanistic link between GLP-1R trafficking and signaling. This study highlights the importance of receptor internalization for full functionality of GLP-1R. Moreover, cross-talk between the two incretin receptors GLP-1R and GIPR is shown to alter receptor trafficking with functional consequences for GLP-1R signaling.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Glucagon/metabolismo , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/patologia , Dinamina I/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Regulação da Expressão Gênica , Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Humanos , Incretinas/metabolismo , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Ligantes , Transporte Proteico , Receptores Acoplados a Proteínas G/genética , Receptores de Glucagon/biossíntese , Transdução de Sinais/genética
4.
Metabolism ; 63(12): 1568-76, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25301258

RESUMO

BACKGROUND: Although it is known that the islet expression of glucagon receptors is increased in type 2 diabetes, its implication for beta-cell function is not known. OBJECTIVE: To determine whether increased beta cell glucagon receptor expression and action influences multiple aspects of beta cell function. MATERIALS/METHODS: Mice with beta cell specific overexpression of the glucagon receptor (RIP-Gcgr) were subjected to intravenous glucose tolerance tests with acute injections of glucagon or GLP-1. Mice were also subjected to intravenous arginine and carbachol tests and insulin secretory responses were evaluated. RESULTS: The specific beta-cell overexpression of glucagon receptors has a complex and diverse consequence with dissociated consequences on beta-cell secretion depending on the stimulatory secretagogue in that whereas the potentiating effects of GLP-1 and arginine on glucose-stimulated insulin secretion were completely lost, the response to the muscarinic receptor agonist carbachol was largely unaffected and the insulin secretory response to glucose was exaggerated. CONCLUSION: This suggests that glucagon receptor overexpression, which is seen in hyperglycemia, may have dissociated consequence on beta cell function in its regulation under fasting, after meal and in response to autonomic nervous activation.


Assuntos
Células Secretoras de Insulina/metabolismo , Receptores de Glucagon/biossíntese , Animais , Arginina/farmacologia , Peso Corporal/efeitos dos fármacos , Carbacol/farmacologia , Glucagon/farmacologia , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Teste de Tolerância a Glucose , Humanos , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Receptores de Glucagon/efeitos dos fármacos , Receptores de Glucagon/genética , Receptores Muscarínicos/efeitos dos fármacos
5.
Eur J Pharmacol ; 741: 311-5, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25179575

RESUMO

The increase in insulin response to oral glucose compared with glucose given by intravenous injection is termed the incretin effect and is mediated by two peptide hormones secreted from the gut in response to nutrient intake: glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). GLP-1 and GIP exert their insulinotropic effects through their respective receptors expressed on pancreatic ß-cells. Both the GLP-1 receptor and the GIP receptor are members of the secretin family of G protein-coupled receptors and couple positively with adenylate cyclase, resulting in an increase in intracellular cAMP. In the present study, we investigated the activity of six previously reported peptide ligands at both the GLP-1 and GIP receptors expressed on HEK-293 cells using a highly sensitive reporter gene assay. GLP-1 and GIP demonstrated almost 100,000-fold selectivity for their respective receptors. Exendin 4 (Ex-4), a long-acting GLP-1 receptor agonist, displayed considerable activity at the GIP receptor. Exendin 9-39 (Ex 9-39) was able to block activity at both the GLP-1 and GIP receptors, and Pro3GIP, a previously-reported GIP receptor antagonist, was shown to act as a partial agonist at the GIP receptor. These data highlight the need for more selective antagonists to study these therapeutically important receptors.


Assuntos
Incretinas/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores dos Hormônios Gastrointestinais/biossíntese , Receptores de Glucagon/biossíntese , Sequência de Aminoácidos , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Humanos , Incretinas/genética , Ligantes , Dados de Sequência Molecular , Fragmentos de Peptídeos/genética , Receptores dos Hormônios Gastrointestinais/antagonistas & inibidores , Receptores de Glucagon/antagonistas & inibidores
6.
Int J Clin Exp Pathol ; 7(7): 4136-42, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25120793

RESUMO

This study is to explore the possible mechanism of ileal interposition (IT) treatment of glycemic control of the type 2 diabetes mellitus (T2DM) by establishing an IT animal model. Twelve T2DM rats (GK rats) of 8-week old were divided into GK IT surgery group (GK-IT) and GK sham group (GK-Sham). Six Wistar rats were used as the non-T2DM sham group (WS-Sham). Enzyme-linked immunosorbent assay was used to detect plasma insulin concentration and fasting pancreas glucagon-like peptide-1 (GLP-1) concentration changes. Homeostasis model assessment of insulin resistance was used to quantitatively measure insulin resistance. Glucagon-like peptide-1 receptor (GLP-1R) expression was detected by Western blotting. IT significantly decreased fasting blood glucose level and the oral glucose tolerance, and reduced insulin resistance of GK rats by increasing GLP-1 concentration and GLP-1R levels. The postoperative pancreatic ß-cell apoptosis rate of GK-Sham group was significantly higher than those in the GK-IT group and the WS-Sham group. IT significantly reduces blood glucose and decreases insulin resistance by up-regulating GLP-1 concentrations and GLP-1R levels, which may contribute to insulin secretion of pancreatic ß-cells and decreases apoptosis of pancreatic ß-cell.


Assuntos
Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 2/cirurgia , Peptídeo 1 Semelhante ao Glucagon/biossíntese , Células Secretoras de Insulina/metabolismo , Receptores de Glucagon/biossíntese , Animais , Glicemia/fisiologia , Western Blotting , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Ensaio de Imunoadsorção Enzimática , Receptor do Peptídeo Semelhante ao Glucagon 1 , Íleo , Resistência à Insulina/fisiologia , Derivação Jejunoileal , Masculino , Ratos , Ratos Wistar , Regulação para Cima
7.
Endocr Pathol ; 25(4): 390-6, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25119061

RESUMO

Neuroendocrine neoplasm (NEN) of the pancreas and gastrointestinal tract is infrequent but often produces hormones to cause distinct clinical features. Glucagon-like peptide-1 receptor (GLP1R) is a G-protein coupled receptor for GLP1, which is cleaved by dipeptidyl peptidase (DPP)-IV, a peptidase that regulates the activity of peptide hormones. Since these molecules are involved in the neuroendocrine function of NEN, they could serve molecular targets for diagnosis and therapy of NEN. However, the expressions of these molecules in NEN are not well studied. We therefore examined the expression of GLP1R and DPP-IV in 22 cases of pancreatic NEN (P-NEN) and 20 cases of gastrointestinal NEN (GI-NEN) by immunostaining. GLP1R was expressed in all eight insulinomas (100 %) but so in only four out of 14 cases (29 %) of non-insulinomas. In contrast to GLP1R, DPP-IV was detected in one out of eight insulinomas (13 %) and in 12 out of 14 cases (86 %) of non-insulinomas. In GI-NEN, GLP1R was negative in all 10 cases of the foregut NEN, whereas it was expressed in all three cases (100 %) of midgut NEN and four out of seven cases (57 %) of hindgut NEN. DPP-IV was expressed in five out of 10 cases (50 %) of the foregut NEN. The expression was detected in two out of three cases (67 %) of midgut NEN and in all seven cases (100 %) of hindgut NEN. In conclusion, we found distinct expression patterns of GLP1R and DPP-IV depending on the neuroendocrine cell types in P-NEN and the anatomical sites in GI-NEN.


Assuntos
Dipeptidil Peptidase 4/biossíntese , Neoplasias Gastrointestinais/metabolismo , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Glucagon/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Dipeptidil Peptidase 4/genética , Feminino , Neoplasias Gastrointestinais/genética , Regulação Neoplásica da Expressão Gênica , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Tumores Neuroendócrinos/genética , Neoplasias Pancreáticas/genética , Receptores de Glucagon/genética , Estudos Retrospectivos , Estatísticas não Paramétricas
8.
Cardiovasc Drugs Ther ; 28(5): 425-32, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25022544

RESUMO

BACKGROUND: Anti-atherosclerotic effects of dipeptidyl peptidase-4 (DPP-4) inhibitors have been shown in many studies. Since inflammation and immune response play a key role in atherogenesis, we examined the effect of DPP-4 inhibitors on the expression of nod-like receptor family, pyrin domain containing 3 (NLRP3) Inflammasome and Interleukin-1beta (IL-1ß) in human macrophages. METHODS AND RESULTS: THP-1 macrophages were incubated with oxidized low density lipoprotein (ox-LDL) with or without DPP-4 inhibitors (sitagliptin and NVPDPP728). The effects of DPP-4 inhibitors on the expression of NLRP3, toll-like receptor 4 (TLR4) and pro-inflammatory cytokine IL-1ß were studied. Both DPP-4 inhibitors induced a significant reduction in NLRP3, TLR4 and IL-1ß expression; concurrently, there was an increase in glucagon like peptide 1 receptor (GLP-1R) expression. Simultaneously, DPP-4 inhibitors reduced phosphorylated-PKC, but not PKA, levels. To determine the role of PKC activation in the effects of DPP-4 inhibitors, cells were treated with PMA- which blocked the effect of DPP-4 inhibitors on NLRP3 and IL-1ß as well as TLR4 and GLP-1R. Over-expression of GLP-1R in macrophages with its agonist liraglutide also blocked the effects of PMA. CONCLUSION: DPP-4 inhibitors suppress NLRP3, TLR4 and IL-1ß in human macrophages through inhibition of PKC activity. This study provides novel insights into the mechanism of inhibition of inflammatory state and immune response in atherosclerosis by DPP-4 inhibitors.


Assuntos
Proteínas de Transporte/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Inflamassomos/biossíntese , Interleucina-1beta/biossíntese , Macrófagos/efeitos dos fármacos , Proteína Quinase C/metabolismo , Receptores de Glucagon/metabolismo , Proteínas de Transporte/biossíntese , Técnicas de Cultura de Células , Proteínas Quinases Dependentes de AMP Cíclico/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Liraglutida , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Nitrilas/antagonistas & inibidores , Nitrilas/farmacologia , Fosforilação/efeitos dos fármacos , Pirazinas/antagonistas & inibidores , Pirazinas/farmacologia , Pirrolidinas/antagonistas & inibidores , Pirrolidinas/farmacologia , Receptores de Glucagon/biossíntese , Transdução de Sinais/efeitos dos fármacos , Fosfato de Sitagliptina , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacologia , Receptor 4 Toll-Like/biossíntese , Triazóis/antagonistas & inibidores , Triazóis/farmacologia
9.
Am J Physiol Gastrointest Liver Physiol ; 307(4): G410-9, 2014 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-24970775

RESUMO

Glucagon-like peptide-2 (GLP-2) has been shown to be effective in patients with short bowel syndrome (SBS), but it is rapidly inactivated by dipeptidyl peptidase IV (DPP4). We used an orally active DPP4 inhibitor (DPP4-I), MK-0626, to determine the efficacy of this approach to promote adaptation after SBS, determined optimal dosing, and identified further functional actions in a mouse model of SBS. Ten-week-old mice underwent a 50% proximal small bowel resection. Dose optimization was determined over a 3-day post-small bowel resection period. The established optimal dose was given for 7, 30, and 90 days and for 7 days followed by a 23-day washout period. Adaptive response was assessed by morphology, intestinal epithelial cell (IEC) proliferation (proliferating cell nuclear antigen), epithelial barrier function (transepithelial resistance), RT-PCR for intestinal transport proteins and GLP-2 receptor, IGF type 1 receptor, and GLP-2 plasma levels. Glucose-stimulated sodium transport was assessed for intestinal absorptive function. Seven days of DPP4-I treatment facilitated an increase in GLP-2 receptor levels, intestinal growth, and IEC proliferation. Treatment led to differential effects over time, with greater absorptive function at early time points and enhanced proliferation at later time points. Interestingly, adaptation continued in the group treated for 7 days followed by a 23-day washout. DPP4-I enhanced IEC proliferative action up to 90 days postresection, but this action seemed to peak by 30 days, as did GLP-2 plasma levels. Thus DPP4-I treatment may prove to be a viable option for accelerating intestinal adaptation with SBS.


Assuntos
Inibidores da Dipeptidil Peptidase IV/farmacologia , Síndrome do Intestino Curto/metabolismo , Adaptação Fisiológica , Amilases/sangue , Animais , Glicemia/metabolismo , Proliferação de Células/efeitos dos fármacos , Dipeptidil Peptidase 4/metabolismo , Modelos Animais de Doenças , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2 , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/citologia , Jejuno/efeitos dos fármacos , Jejuno/ultraestrutura , Masculino , Camundongos , Receptor IGF Tipo 1/biossíntese , Receptores de Glucagon/biossíntese , Transportador 1 de Glucose-Sódio/biossíntese , Triazóis/uso terapêutico , Regulação para Cima
10.
Int J Mol Med ; 34(2): 475-82, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24866833

RESUMO

Glucose-dependent insulinotropic polypeptide receptor (GIPR) and glucagon-like peptide-1 receptor (GLP­1R) are incretin receptors that play important roles in regulating insulin secretion from pancreatic ß cells. Incretin receptors are also thought to play a potential role in bone metabolism. Osteoblasts in animals and humans express GIPR; however, the presence of GLP-1R in these cells has not been reported to date. Thus, the aim of this study was to determine whether GLP-1R and GIPR are expressed in osteoblastic cells, and whether their expression levels are regulated by the extracellular glucose concentration. Mouse osteoblastic MC3T3-E1 cells were cultured in medium containing normal (5.6 mM) or high (10, 20 or 30 mM) glucose concentrations, with or without bone morphogenetic protein-2 (BMP-2). RT-PCR, western blot analysis and immunofluorescence were carried out to determine GIPR and GLP-1R mRNA and protein expression levels. Cell proliferation was also assessed. The GLP-1R and GIPR mRNA expression levels were higher in the MC3T3-E1 cells cultured in medium containing high glucose concentrations with BMP-2 compared with the cells cultured in medium containing normal glucose concentrations with or without BMP-2. GLP-1R protein expression increased following culture in high-glucose medium with BMP-2 compared with culture under normal glucose conditions. However, the cellular localization of GLP-1R was not affected by either glucose or BMP-2. In conclusion, our data demonstrate that the expression of GLP-1R and GIPR is regulated by glucose concentrations in MC3T3-E1 cells undergoing differentiation induced by BMP-2. Our results reveal the potential role of incretins in bone metabolism.


Assuntos
Glucose/metabolismo , Insulina/metabolismo , Receptores dos Hormônios Gastrointestinais/biossíntese , Receptores de Glucagon/biossíntese , Animais , Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular/genética , Linhagem Celular , Polipeptídeo Inibidor Gástrico , Regulação da Expressão Gênica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Camundongos , Osteoblastos/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Glucagon/metabolismo
12.
Mol Biol Rep ; 41(6): 3853-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24549718

RESUMO

High mobility group box 1 protein (HMGB1) plays an important role in myocardial ischemia and reperfusion (I/R) injury. Preconditioning of exendin-4 (Ex), a glucagon-like peptide-1 receptor agonist, has been reported to attenuate myocardial I/R injury. The current study investigated whether Ex postconditioning also attenuated myocardial I/R injury and the potential mechanisms. Anesthetized male rats were subjected to ischemia for 30 min and treated with Ex (5 µg/kg, i.v.) 5 min before reperfusion, in the absence and/or presence of exendin (9-39) (an antagonist of glucagon-like peptide-1 receptor, 5 µg/kg, i.v.), followed by reperfusion for 4 h. Lactate dehydrogenase (LDH), creatine kinase (CK), tumor necrosis factor-α, interleukin-6, and infarct size were measured. HMGB1 expression was assessed by immunoblotting. Postconditioning with Ex significantly decreased infarct size and levels of LDH and CK after 4 h reperfusion (all p < 0.05). Ex also significantly inhibited the increase in malondialdehyde level and decreased the level of superoxide dismutase (both p < 0.05). In addition, the increase in HMGB1 expression induced by I/R was significantly attenuated by Ex postconditioning. Administration of exendin (9-39) abolished the protective effect of Ex postconditioning (all p < 0.05). The present study suggests that Ex postconditioning may attenuate myocardial I/R injury, which may in turn be associated with inhibiting inflammation.


Assuntos
Inflamação/tratamento farmacológico , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Peptídeos/administração & dosagem , Receptores de Glucagon/agonistas , Peçonhas/administração & dosagem , Animais , Creatina Quinase/biossíntese , Exenatida , Regulação da Expressão Gênica/efeitos dos fármacos , Receptor do Peptídeo Semelhante ao Glucagon 1 , Proteína HMGB1/biossíntese , Proteína HMGB1/metabolismo , Humanos , Inflamação/genética , Inflamação/patologia , Interleucina-6/biossíntese , L-Lactato Desidrogenase/biossíntese , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/patologia , Fragmentos de Peptídeos/administração & dosagem , Peptídeos/metabolismo , Ratos , Receptores de Glucagon/biossíntese , Fator de Necrose Tumoral alfa/biossíntese , Peçonhas/metabolismo
13.
Peptides ; 52: 134-42, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24406898

RESUMO

Glucagon-like peptide-1 (GLP-1) has been proved to have effects of anti-hyperglycemia and ß-cell preservation. However, it is still unclear whether there are differences between early and late GLP-1 intervention in type 2 diabetes mellitus (T2DM). We divided the mice into 5 groups: early treated group (n=7, 8-week old, fasting glucose>10mmol/l), late treated group (n=7, 10-week old, fasting glucose>20mmol/l), early control group (n=7), late control group (n=7) and wild type group (n=7). Treated group was injected with liraglutide (a GLP-1 analog) 300µg/kg bid for 4 weeks, while control group was given saline at the same time. The results showed that compared with control group, food intake and body weight gain were reduced in both early and late treated group (p<0.05), and there was no significance between the two treated groups. Early liraglutide intervention showed better improvements in glucose control, acute insulin response to glucose (AIRg) and disposition index (before vs. after treatment, AIRg 1.01±0.53 vs. 2.98±0.63, disposition index 10.81±0.89 vs. 27.4±2.15) than late intervention (AIRg 0.99±0.02 vs. 1.41±0.32, disposition index 3.47±0.38 vs. 6.43±1.62, p=0.001). The histopathology of the pancreas showed the estimated ß-cell mass (BCM) was increased more in early treated group than that in late one (0.03 vs. 0.01g). Expressions of the proliferation related genes PDX-1, MafA and GLP-1 receptor (GLP-1R) in early treated group were 1.81, 2.57 and 1.59 times as much as that in late treated group. In conclusion, early liraglutide intervention was better in glucose control, ß-cell function improvement and ß-cell mass preservation.


Assuntos
Índice de Massa Corporal , Ingestão de Alimentos/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Glucose/metabolismo , Hipoglicemiantes/farmacologia , Células Secretoras de Insulina/metabolismo , Animais , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Proteínas de Homeodomínio/biossíntese , Células Secretoras de Insulina/patologia , Liraglutida , Fatores de Transcrição Maf Maior/biossíntese , Masculino , Camundongos , Receptores de Glucagon/biossíntese , Transativadores/biossíntese
14.
Pancreas ; 43(1): 1-6, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24326362

RESUMO

OBJECTIVES: Glucagon-like peptide 1 (GLP-1) interacts with its specific high-affinity receptor, glucagon-like peptide 1 receptor (GLP-1R), and induces cellular growth and inhibition of apoptosis in pancreatic ß cells. The aim of this study was to investigate the significance of GLP-1R expression in pancreatic neuroendocrine tumors (PNETs). METHODS: Glucagon-like peptide 1 receptor expression was semiquantitatively evaluated by immunohistochemical staining in 50 resected PNETs, and the correlation between the GLP-1R expression and clinicopathologic features was investigated. RESULTS: There were 23 PNETs with positive expression and 27 PNETs with negative expression of GLP-1R. Positive expression of GLP-1R was more frequently observed in insulinoma than in gastrinoma and nonfunctioning tumor (P < 0.05). Although expression status of GLP-1R did not affect the prognosis of the patients with PNETs (P = 0.82), most of the metastatic sites such as lymph node and liver showed positive staining for GLP-1R (8 of 11 PNETs, 73%). CONCLUSIONS: Glucagon-like peptide 1 receptor would be a diagnostic marker of insulinoma and might become a molecular target for treatment of metastatic PNETs and hormonal regulation of insulin.


Assuntos
Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Receptores de Glucagon/biossíntese , Adulto , Idoso , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Análise de Sobrevida , Adulto Jovem
15.
Bioconjug Chem ; 25(1): 171-7, 2014 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-24328216

RESUMO

The ability to reliably identify pancreatic ß-cells would have far reaching implications for a greater understanding of ß-cell biology, measurement of ß-cell mass in diabetes, islet transplantation, and drug development. The glucagon-like peptide-1 receptor (GLP1R) is highly expressed on the surface of insulin producing pancreatic ß-cells. Using systematic modifications of the GLP1R ligand, exendin-4, we screened over 25 compounds and identified a palette of fluorescent exendin-4 with high GLP1R binding affinity. We show considerable differences in affinity, as well as utility of the top candidates for flow cytometry and microscopy of ß-cells. Some of the developed compounds should be particularly useful for basic and translational ß-cell research.


Assuntos
Fluorescência , Células Secretoras de Insulina/citologia , Peptídeos/química , Peçonhas/química , Animais , Células Cultivadas , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Células HEK293 , Humanos , Células Secretoras de Insulina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Estrutura Molecular , Peptídeos/síntese química , Receptores de Glucagon/biossíntese , Receptores de Glucagon/química , Peçonhas/síntese química
16.
Annu Rev Physiol ; 76: 561-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24161075

RESUMO

Glucagon-like peptide-2 (GLP-2) is a 33-amino-acid proglucagon-derived peptide secreted from enteroendocrine L cells. GLP-2 circulates at low basal levels in the fasting period, and plasma levels rise rapidly after food ingestion. Renal clearance and enzymatic inactivation control the elimination of bioactive GLP-2. GLP-2 increases mesenteric blood flow and activates proabsorptive pathways in the gut, facilitating nutrient absorption. GLP-2 also enhances gut barrier function and induces proliferative and cytoprotective pathways in the small bowel. The actions of GLP-2 are transduced via a single G protein-coupled receptor (GLP-2R), expressed predominantly within the gastrointestinal tract. Disruption of GLP-2R signaling increases susceptibility to gut injury and impairs the adaptive mucosal response to refeeding. Sustained augmentation of GLP-2R signaling reduces the requirement for parenteral nutrition in human subjects with short-bowel syndrome. Hence GLP-2 integrates nutrient-derived signals to optimize mucosal integrity and energy absorption.


Assuntos
Peptídeo 2 Semelhante ao Glucagon/farmacologia , Peptídeo 2 Semelhante ao Glucagon/fisiologia , Animais , Gastroenteropatias/fisiopatologia , Motilidade Gastrointestinal/fisiologia , Trato Gastrointestinal/irrigação sanguínea , Trato Gastrointestinal/fisiologia , Receptor do Peptídeo Semelhante ao Glucagon 2 , Humanos , Absorção Intestinal , Isquemia/fisiopatologia , Lesões por Radiação/fisiopatologia , Receptores de Glucagon/biossíntese , Receptores de Glucagon/efeitos dos fármacos , Receptores de Glucagon/metabolismo , Síndrome do Intestino Curto/fisiopatologia , Transdução de Sinais
17.
J Neurochem ; 128(3): 459-71, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24112036

RESUMO

Glucagon-like peptide 1 (GLP-1) is a growth factor that has demonstrated neuroprotective properties in a range of studies. In an APPswe/PS1ΔE9 mouse model of Alzheimer's disease (AD), we previously found protective effects on memory formation, synaptic plasticity, synapse survival and a reduction of amyloid synthesis and plaque load in the brain. Here, we analyse the neuroprotective properties of the GLP-1 analogue liraglutide in human neuroblastoma cell line SH-SY5Y during methyl glyoxal stress. We show for the first time that cell viability was enhanced by liraglutide (XTT assay) in a dose-dependent way, while cytotoxicity (LDH assay) and apoptosis were reduced. Expression of the pro-survival Mcl1 signaling protein was increased, as was activation of cell survival kinases Akt, MEK1/2 and the transcription factor p90RSK. Liraglutide also decreased pro-apoptotic Bax and Bik expression. In addition, the membrane potential and the influx of calcium into the cell were enhanced by liraglutide. GLP-1 receptor expression was also increased by the drug. The results demonstrate a range of growth factor-related cytoprotective processes induced by liraglutide, which is currently on the market as a treatment for type 2 diabetes (Victoza®). It is also tested in clinical trials in patients with Alzheimer disease.


Assuntos
Apoptose/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Hipoglicemiantes/farmacologia , Fármacos Neuroprotetores , Aldeído Pirúvico/toxicidade , Estresse Fisiológico/efeitos dos fármacos , Proteínas Reguladoras de Apoptose/metabolismo , Western Blotting , Cálcio/metabolismo , Caspase 3/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Imuno-Histoquímica , Liraglutida , Potenciais da Membrana , Proteínas de Membrana/metabolismo , Proteínas Mitocondriais , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteína Oncogênica v-akt/metabolismo , Receptores de Glucagon/biossíntese , Receptores de Glucagon/genética , Proteína X Associada a bcl-2/metabolismo
18.
Diabetes ; 63(4): 1224-33, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24296712

RESUMO

GLP-1 is an intestinal hormone with widespread actions on metabolism. Therapies based on GLP-1 are highly effective because they increase glucose-dependent insulin secretion in people with type 2 diabetes, but many reports suggest that GLP-1 has additional beneficial or, in some cases, potentially dangerous actions on other tissues, including the heart, vasculature, exocrine pancreas, liver, and central nervous system. Identifying which tissues express the GLP-1 receptor (GLP1R) is critical for the development of GLP-1-based therapies. Our objective was to use a method independent of GLP1R antibodies to identify and characterize the targets of GLP-1 in mice. Using newly generated glp1r-Cre mice crossed with fluorescent reporter strains, we show that major sites of glp1r expression include pancreatic ß- and δ-cells, vascular smooth muscle, cardiac atrium, gastric antrum/pylorus, enteric neurones, and vagal and dorsal root ganglia. In the central nervous system, glp1r-fluorescent cells were abundant in the area postrema, arcuate nucleus, paraventricular nucleus, and ventromedial hypothalamus. Sporadic glp1r-fluorescent cells were found in pancreatic ducts. No glp1r-fluorescence was observed in ventricular cardiomyocytes. Enteric and vagal neurons positive for glp1r were activated by GLP-1 and may contribute to intestinal and central responses to locally released GLP-1, such as regulation of intestinal secretomotor activity and appetite.


Assuntos
Receptores de Glucagon/biossíntese , Animais , Sistema Nervoso Central/citologia , Sistema Nervoso Central/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1 , Átrios do Coração/citologia , Átrios do Coração/metabolismo , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/metabolismo , Camundongos , Camundongos Transgênicos , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo
19.
Endocrinology ; 154(2): 666-74, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23307791

RESUMO

Ghrelin is a stomach-derived orexigenic hormone whose levels in circulation are altered by energy availability. Like ghrelin, the glucotropic hormone glucagon increases in the fasting state and serves to normalize energy levels. We hypothesized that glucagon can directly stimulate stomach ghrelin production. To verify this hypothesis, we used a primary culture of dispersed rat stomach cells. We first demonstrated that stomach ghrelin cells express the glucagon receptor (GluR). Glucagon (1-100 nM) significantly stimulated ghrelin secretion and proghrelin mRNA expression, and co-incubation with a GluR inhibitor prevented glucagon's action. The MAP kinase inhibitor (PD98058) reduced the glucagon-stimulated ghrelin secretion and proghrelin mRNA expression. Furthermore, glucagon treatment increased the phosphorylation of ERK1/2. Glucagon also increased intracellular cAMP levels, and inhibition of adenylate cyclase reduced glucagon's effect on ghrelin secretion. Surprisingly, inhibiting protein kinase A (PKA) (using H89 and phosphorothioate [Rp]-cAMP) did not prevent glucagon-stimulated ghrelin secretion. Instead, inhibiting the exchange protein activated by cAMP (EPAC) with Brefeldin-A was able to significantly reduce glucagon-stimulated ghrelin secretion. Furthermore, the EPAC agonist (8-pCPT) significantly stimulated ghrelin secretion. Depleting endoplasmic reticulum calcium stores or blocking voltage-dependant calcium channels prevented glucagon stimulated ghrelin secretion. Finally, co-incubation with the sympathetic neurotransmitter norepinephrine potentiated the glucagon stimulation of ghrelin secretion. Our findings are the first to show a direct link between glucagon and stomach ghrelin production and secretion and highlight the role of MAPK, the PKA-independent EPAC pathway, and the synergy between norepinephrine and glucagon in ghrelin release.


Assuntos
Grelina/metabolismo , Glucagon/farmacologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , MAP Quinase Quinase Quinases/metabolismo , Norepinefrina/farmacologia , 8-Bromo Monofosfato de Adenosina Cíclica/análogos & derivados , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Cálcio/metabolismo , Caproatos/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Sinergismo Farmacológico , Feminino , MAP Quinase Quinase Quinases/antagonistas & inibidores , Masculino , RNA Mensageiro/metabolismo , Ratos , Receptores de Glucagon/biossíntese , Estômago/citologia , Estômago/efeitos dos fármacos
20.
Toxicol Appl Pharmacol ; 264(2): 274-83, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22925809

RESUMO

The glucagon-like peptide 1 receptor (GLP1R) plays a critical role in glucose metabolism and has become an important target for a growing class of drugs designed to treat type 2 diabetes. In vitro studies were designed to investigate the effect of the GLP1R agonist, exenatide (Ex4), in "on-target" RIN-5mF (islet) cells as well as in "off-target" AR42J (acinar) and DSL-6A/C1 (ductal) cells in a diabetic environment. Ex4 increased islet cell proliferation but did not affect acinar cells or ductal cells at relevant concentrations. A high caloric, high fat diet is a risk factor for impaired glucose tolerance and type-2 diabetes. An in vitro Oleic acid (OA) model was used to investigate the effect of Ex4 in a high calorie, high fat environment. At 0.1 and 0.4mM, OA mildly decreased the proliferation of all pancreatic cell types. Ex4 did not potentiate the inhibitory effect of OA on cell proliferation. Akt phosphorylation in response to Ex4 was diminished in OA-treated ductal cells. GLP1R protein detected by western blot was time and concentration dependently decreased after glucose stimulation in OA-treated ductal cells. In ductal cells, OA treatment altered the intracellular localization of GLP1R and its co-localization with early endosome and recycling endosomes. Chloroquine (lysosomal inhibitor), N-acetyl-l-cysteine (reactive oxygen species scavenger) and wortmannin (a phosphatidylinositol-3-kinase inhibitor), fully or partially, rescued GLP1R protein in OA-pretreated, glucose-stimulated ductal cells. The impact of altered regulation on phenotype/function is presently unknown. However, these data suggest that GLP1R regulation in ductal cells can be altered by a high fat, high calorie environment.


Assuntos
Glucose/farmacologia , Ácido Oleico/farmacologia , Ductos Pancreáticos/efeitos dos fármacos , Ductos Pancreáticos/metabolismo , Receptores de Glucagon/biossíntese , Células Acinares/efeitos dos fármacos , Células Acinares/metabolismo , Animais , Western Blotting , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Endossomos/efeitos dos fármacos , Endossomos/metabolismo , Exenatida , Imunofluorescência , Receptor do Peptídeo Semelhante ao Glucagon 1 , Ilhotas Pancreáticas/efeitos dos fármacos , Ilhotas Pancreáticas/metabolismo , Microscopia Confocal , Proteína Oncogênica v-akt/metabolismo , Neoplasias Pancreáticas/metabolismo , Peptídeos/farmacologia , Fosforilação , Ratos , Receptores de Glucagon/agonistas , Estimulação Química , Vacúolos/efeitos dos fármacos , Peçonhas/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA