Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Br J Pharmacol ; 179(9): 1998-2015, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34855984

RESUMO

BACKGROUND: Glucagon-like peptide-2 (GLP-2) is a pro-glucagon-derived hormone secreted from intestinal enteroendocrine L cells with actions on gut and bones. GLP-2(1-33) is cleaved by DPP-4, forming GLP-2(3-33), having low intrinsic activity and competitive antagonism properties at GLP-2 receptors. We created radioligands based on these two molecules. EXPERIMENTAL APPROACH: The methionine in position 10 of GLP-2(1-33) and GLP-2(3-33) was substituted with tyrosine (M10Y) enabling oxidative iodination, creating [125 I]-hGLP-2(1-33,M10Y) and [125 I]-hGLP-2(3-33,M10Y). Both were characterized by competition binding, on-and-off-rate determination and receptor activation. Receptor expression was determined by target-tissue autoradiography and immunohistochemistry. KEY RESULTS: Both M10Y-substituted peptides induced cAMP production via the GLP-2 receptor comparable to the wildtype peptides. GLP-2(3-33,M10Y) maintained the antagonistic properties of GLP-2(3-33). However, hGLP-2(1-33,M10Y) had lower arrestin recruitment than hGLP-2(1-33). High affinities for the hGLP-2 receptor were observed using [125 I]-hGLP-2(1-33,M10Y) and [125 I]-hGLP-2(3-33,M10Y) with KD values of 59.3 and 40.6 nM. The latter (with antagonistic properties) had higher Bmax and faster on and off rates compared to the former (full agonist). Both bound the hGLP-1 receptor with low affinity (Ki of 130 and 330 nM, respectively). Autoradiography in wildtype mice revealed strong labelling of subepithelial myofibroblasts, confirmed by immunohistochemistry using a GLP-2 receptor specific antibody that in turn was confirmed in GLP-2 receptor knock-out mice. CONCLUSION AND IMPLICATIONS: Two new radioligands with different binding kinetics, one a full agonist and the other a weak partial agonist with antagonistic properties were developed and subepithelial myofibroblasts identified as a major site for GLP-2 receptor expression.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 2 , Peptídeos , Animais , Ligação Competitiva , Receptor do Peptídeo Semelhante ao Glucagon 2/agonistas , Receptor do Peptídeo Semelhante ao Glucagon 2/antagonistas & inibidores , Humanos , Camundongos , Fragmentos de Peptídeos/metabolismo , Peptídeos/farmacologia
2.
Mol Metab ; 42: 101080, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32937194

RESUMO

OBJECTIVE: Glucagon is well known to regulate blood glucose but may be equally important for amino acid metabolism. Plasma levels of amino acids are regulated by glucagon-dependent mechanism(s), while amino acids stimulate glucagon secretion from alpha cells, completing the recently described liver-alpha cell axis. The mechanisms underlying the cycle and the possible impact of hepatic steatosis are unclear. METHODS: We assessed amino acid clearance in vivo in mice treated with a glucagon receptor antagonist (GRA), transgenic mice with 95% reduction in alpha cells, and mice with hepatic steatosis. In addition, we evaluated urea formation in primary hepatocytes from ob/ob mice and humans, and we studied acute metabolic effects of glucagon in perfused rat livers. We also performed RNA sequencing on livers from glucagon receptor knock-out mice and mice with hepatic steatosis. Finally, we measured individual plasma amino acids and glucagon in healthy controls and in two independent cohorts of patients with biopsy-verified non-alcoholic fatty liver disease (NAFLD). RESULTS: Amino acid clearance was reduced in mice treated with GRA and mice lacking endogenous glucagon (loss of alpha cells) concomitantly with reduced production of urea. Glucagon administration markedly changed the secretion of rat liver metabolites and within minutes increased urea formation in mice, in perfused rat liver, and in primary human hepatocytes. Transcriptomic analyses revealed that three genes responsible for amino acid catabolism (Cps1, Slc7a2, and Slc38a2) were downregulated both in mice with hepatic steatosis and in mice with deletion of the glucagon receptor. Cultured ob/ob hepatocytes produced less urea upon stimulation with mixed amino acids, and amino acid clearance was lower in mice with hepatic steatosis. Glucagon-induced ureagenesis was impaired in perfused rat livers with hepatic steatosis. Patients with NAFLD had hyperglucagonemia and increased levels of glucagonotropic amino acids, including alanine in particular. Both glucagon and alanine levels were reduced after diet-induced reduction in Homeostatic Model Assessment for Insulin Resistance (HOMA-IR, a marker of hepatic steatosis). CONCLUSIONS: Glucagon regulates amino acid metabolism both non-transcriptionally and transcriptionally. Hepatic steatosis may impair glucagon-dependent enhancement of amino acid catabolism.


Assuntos
Aminoácidos/metabolismo , Fígado Gorduroso/fisiopatologia , Glucagon/metabolismo , Adulto , Animais , Glicemia/metabolismo , Fígado Gorduroso/metabolismo , Feminino , Glucagon/fisiologia , Células Secretoras de Glucagon/metabolismo , Glucose/metabolismo , Hepatócitos/metabolismo , Humanos , Insulina/metabolismo , Resistência à Insulina/fisiologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Hepatopatia Gordurosa não Alcoólica/metabolismo , Ratos , Ratos Wistar , Receptores de Glucagon/antagonistas & inibidores , Receptores de Glucagon/metabolismo , Ureia/metabolismo
3.
Int J Obes (Lond) ; 44(9): 1859-1871, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32015474

RESUMO

OBJECTIVES: Gastrointestinal hormones contribute to the beneficial effects of Roux-en-Y gastric bypass surgery (RYGB) on glycemic control. Secretin is secreted from duodenal S cells in response to low luminal pH, but it is unknown whether its secretion is altered after RYGB and if secretin contributes to the postoperative improvement in glycemic control. We hypothesized that secretin secretion increases after RYGB as a result of the diversion of nutrients to more distal parts of the small intestine, and thereby affects islet hormone release. METHODS: A specific secretin radioimmunoassay was developed, evaluated biochemically, and used to quantify plasma concentrations of secretin in 13 obese individuals before, 1 week after, and 3 months after RYGB. Distribution of secretin and its receptor was assessed by RNA sequencing, mass-spectrometry and in situ hybridization in human and rat tissues. Isolated, perfused rat intestine and pancreas were used to explore the molecular mechanism underlying glucose-induced secretin secretion and to study direct effects of secretin on glucagon, insulin, and somatostatin secretion. Secretin was administered alone or in combination with GLP-1 to non-sedated rats to evaluate effects on glucose regulation. RESULTS: Plasma postprandial secretin was more than doubled in humans after RYGB (P < 0.001). The distal small intestine harbored secretin expressing cells in both rats and humans. Glucose increased the secretion of secretin in a sodium-glucose cotransporter dependent manner when administered to the distal part but not into the proximal part of the rat small intestine. Secretin stimulated somatostatin secretion (fold change: 1.59, P < 0.05) from the perfused rat pancreas but affected neither insulin (P = 0.2) nor glucagon (P = 0.97) secretion. When administered to rats in vivo, insulin secretion was attenuated and glucagon secretion increased (P = 0.04), while blood glucose peak time was delayed (from 15 to 45 min) and gastric emptying time prolonged (P = 0.004). CONCLUSIONS: Glucose-sensing secretin cells located in the distal part of the small intestine may contribute to increased plasma concentrations observed after RYGB. The metabolic role of the distal S cells warrants further studies.


Assuntos
Células Enteroendócrinas , Derivação Gástrica , Glucose/metabolismo , Intestino Delgado/citologia , Animais , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/fisiologia , Masculino , Período Pós-Prandial/fisiologia , Ratos , Ratos Wistar
4.
J Clin Endocrinol Metab ; 104(12): 6403-6416, 2019 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-31276156

RESUMO

CONTEXT: After Roux-en-Y gastric bypass (RYGB) surgery, postprandial plasma glucagon concentrations have been reported to increase. This occurs despite concomitant improved glucose tolerance and increased circulating plasma concentrations of insulin and the glucagon-inhibiting hormone glucagon-like peptide 1 (GLP-1). OBJECTIVE: To investigate whether RYGB-induced hyperglucagonemia may be derived from the gut. DESIGN AND SETTING: Substudy of a prospective cross-sectional study at a university hospital in Copenhagen, Denmark. PARTICIPANTS: Morbidly obese individuals undergoing RYGB (n = 8) with or without type 2 diabetes. INTERVENTIONS: Three months before and after RYGB, participants underwent upper enteroscopy with retrieval of gastrointestinal mucosal biopsy specimens. Mixed-meal tests were performed 1 week and 3 months before and after RYGB. MAIN OUTCOME MEASURES: The 29-amino acid glucagon concentrations in plasma and in mucosal gastrointestinal biopsy specimens were assessed using mass spectrometry-validated immunoassays, and a new monoclonal antibody reacting with immunoreactive glucagon was used for immunohistochemistry. RESULTS: Postprandial plasma concentrations of glucagon after RYGB were increased. Expression of the glucagon gene in the small intestine increased after surgery. Glucagon was identified in the small-intestine biopsy specimens obtained after, but not before, RYGB. Immunohistochemically, mucosal biopsy specimens from the small intestine harbored cells costained for GLP-1 and immunoreactive glucagon. CONCLUSION: Increased concentrations of glucagon were observed in small-intestine biopsy specimens and postprandially in plasma after RYGB. The small intestine harbored cells immunohistochemically costaining for GLP-1 and glucagon-like immunoreactivity after RYGB. Glucagon derived from small-intestine enteroendocrine l cells may contribute to postprandial plasma concentrations of glucagon after RYGB.


Assuntos
Diabetes Mellitus Tipo 2/fisiopatologia , Derivação Gástrica/métodos , Peptídeo 1 Semelhante ao Glucagon/sangue , Glucagon/sangue , Insulina/sangue , Intestinos/fisiologia , Obesidade Mórbida/sangue , Adolescente , Adulto , Enteroscopia de Balão , Biomarcadores/sangue , Glicemia/análise , Estudos de Casos e Controles , Estudos Transversais , Feminino , Seguimentos , Polipeptídeo Inibidor Gástrico/sangue , Hemoglobinas Glicadas/análise , Humanos , Masculino , Refeições , Pessoa de Meia-Idade , Obesidade Mórbida/complicações , Obesidade Mórbida/cirurgia , Período Pós-Prandial , Prognóstico , Estudos Prospectivos , Adulto Jovem
5.
J Endocrinol ; 239(1): 73­80, 2018 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-30307155

RESUMO

Fibroblast growth factor 21 (FGF21) is a metabolic regulator of energy and lipid metabolism. FGF21 is highly expressed in liver while FGF21 receptors (beta-klotho (KLB) and FGFR1c) are highly expressed in white adipose tissues (WATs). Plasma FGF21 has been shown to be increased after 7­10 days of fasting but oppositely plasma FGF21 is also increased in obesity. The aim of this study was to measure the effect of 60 h of fasting on plasma FGF21 levels in obese and lean subjects and to determine the gene expression of KLB and FGFR1c in the subcutaneous WAT before, during and after 60 h of fasting. Eight obese (BMI >30 kg/m2) and seven lean subjects (BMI <25 kg/m2) were fasted for 60 h and blood samples were taken at time 0 and after 12, 36 and 60 h of fasting. A biopsy from the subcutaneous WAT was taken at time 0, 12 and 60 h of fasting. FGF21 was measured in plasma by an ELISA and mRNA expression of KLB and FGFR1c was measured in WAT by quantitative PCR (qPCR). The fast significantly decreased plasma FGF21 in obese subjects while no change in plasma FGF21 was observed in lean subjects. Interestingly, KLB was significantly decreased in WAT in response to fasting in both lean and obese subjects indicating a potential important adaptive regulation of KLB in response to fasting.


Assuntos
Tecido Adiposo Branco/metabolismo , Jejum/sangue , Fatores de Crescimento de Fibroblastos/sangue , Obesidade/sangue , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Adulto , Humanos , Proteínas Klotho , Masculino , Proteínas de Membrana/metabolismo
6.
Endocrinology ; 155(4): 1280-90, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24467746

RESUMO

Glucagon-like peptide 1 (GLP-1) analogs are increasingly being used in the treatment of type 2 diabetes. It is clear that these drugs lower blood glucose through an increase in insulin secretion and a lowering of glucagon secretion; in addition, they lower body weight and systolic blood pressure and increase heart rate. Using a new monoclonal antibody for immunohistochemistry, we detected GLP-1 receptor (GLP-1R) in important target organs in humans and monkeys. In the pancreas, GLP-1R was predominantly localized in ß-cells with a markedly weaker expression in acinar cells. Pancreatic ductal epithelial cells did not express GLP-1R. In the kidney and lung, GLP-1R was exclusively expressed in smooth muscle cells in the walls of arteries and arterioles. In the heart, GLP-1R was localized in myocytes of the sinoatrial node. In the gastrointestinal tract, the highest GLP-1R expression was seen in the Brunner's gland in the duodenum, with lower level expression in parietal cells and smooth muscle cells in the muscularis externa in the stomach and in myenteric plexus neurons throughout the gut. No GLP-1R was seen in primate liver and thyroid. GLP-1R expression seen with immunohistochemistry was confirmed by functional expression using in situ ligand binding with (125)I-GLP-1. In conclusion, these results give important new insight into the molecular mode of action of GLP-1 analogs by identifying the exact cellular localization of GLP-1R.


Assuntos
Anticorpos Monoclonais/química , Insulina/metabolismo , Receptores de Glucagon/metabolismo , Animais , Pressão Sanguínea , Peso Corporal , Linhagem Celular , Cricetinae , Duodeno/metabolismo , Exenatida , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Peptídeo 1 Semelhante ao Glucagon/química , Receptor do Peptídeo Semelhante ao Glucagon 1 , Haplorrinos , Frequência Cardíaca , Humanos , Secreção de Insulina , Ligantes , Liraglutida , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Peptídeos/química , Ligação Proteica , Distribuição Tecidual , Transfecção , Peçonhas/química
7.
J Immunol ; 184(8): 4431-9, 2010 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-20212099

RESUMO

Adenoviral vectors have been widely used for experimental gene therapy and vaccination, yet there is a surprising lack of knowledge connecting the route and dose of adenovirus administration to the induced transgene-specific immune response. We have recently demonstrated polyfunctional CD8(+) T cells and protective memory responses using adenoviral vectors, which seem to contrast with recent reports suggesting that an exhausted CD8(+) T cell phenotype is induced by inoculation with adenoviral vectors. Accordingly, we investigated the route and dose interrelationship for transgene-specific CD8(+) T cells using adenoviral vectors encoding beta-galactosidase applied either s.c. or i.v. Irrespective of the route of inoculation, most of the adenoviral inoculum was found to disseminate systemically as the dose was raised beyond 10(9) particles. The number of transgene-specific CD8(+) T cells correlated positively with dissemination, whereas the functional capacity of the generated T cells correlated inversely with vector dissemination. A comparison of the immune response to s.c. or i.v. administration at moderate doses revealed that inoculation by both routes induced a transient peak of IFN-gamma-producing CD8(+) T cells 2 to 3 wk postinfection, but following i.v. administration, these cells were only detected in the liver. Two to four months after systemic, but not peripheral, immunization, dysfunctional transgene-specific CD8(+) T cells impaired in both cytokine production and important in vivo effector functions, accumulated in the spleen. These findings indicate that the localization of the adenoviral inoculum and not the total Ag load determines the quality of the CD8(+) T cell response induced with adenoviral vaccines.


Assuntos
Adenovírus Humanos/genética , Adenovírus Humanos/imunologia , Linfócitos T CD8-Positivos/imunologia , Testes Imunológicos de Citotoxicidade/métodos , Vetores Genéticos/imunologia , Transgenes/imunologia , Carga Viral/imunologia , Infecções por Adenovirus Humanos/imunologia , Infecções por Adenovirus Humanos/prevenção & controle , Animais , Linfócitos T CD8-Positivos/virologia , Testes Imunológicos de Citotoxicidade/tendências , Relação Dose-Resposta Imunológica , , Vetores Genéticos/administração & dosagem , Vetores Genéticos/normas , Membro Posterior , Memória Imunológica/genética , Injeções Intravenosas , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Vaccinia virus/enzimologia , Vaccinia virus/genética , Vaccinia virus/imunologia , Carga Viral/normas , beta-Galactosidase/administração & dosagem , beta-Galactosidase/genética , beta-Galactosidase/imunologia
8.
J Immunol ; 180(5): 3339-46, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18292559

RESUMO

The ideal vaccine induces a potent protective immune response, which should be rapidly induced, long-standing, and of broad specificity. Recombinant adenoviral vectors induce potent Ab and CD8+ T cell responses against transgenic Ags within weeks of administration, and they are among the most potent and versatile Ag delivery vehicles available. However, the impact of chronic infections like HIV and hepatitis C virus underscore the need for further improvements. In this study, we show that the protective immune response to an adenovirus-encoded vaccine Ag can be accelerated, enhanced, broadened, and prolonged by tethering of the rAg to the MHC class II-associated invariant chain (Ii). Thus, adenovirus-vectored vaccines expressing lymphocytic choriomeningitis virus (LCMV)-derived glycoprotein linked to Ii increased the CD4+ and CD8+ T cell stimulatory capacity in vitro and in vivo. Furthermore, mice vaccinated with a single dose of adenovirus-expressing LCMV-derived glycoprotein linked to Ii were protected against lethal virus-induced choriomeningitis, lethal challenge with strains mutated in immunodominant T cell epitopes, and systemic infection with a highly invasive strain. In therapeutic tumor vaccination, the vaccine was as efficient as live LCMV. In comparison, animals vaccinated with a conventional adenovirus vaccine expressing unmodified glycoprotein were protected against systemic infection, but only temporarily against lethal choriomeningitis, and this vaccine was less efficient in tumor therapy.


Assuntos
Adenoviridae/imunologia , Antígenos de Diferenciação de Linfócitos B/administração & dosagem , Antígenos de Diferenciação de Linfócitos B/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Antígenos de Histocompatibilidade Classe II/administração & dosagem , Antígenos de Histocompatibilidade Classe II/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Vacinas Virais/imunologia , Adenoviridae/genética , Animais , Antígenos de Diferenciação de Linfócitos B/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD8-Positivos/citologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Células Cultivadas , Técnicas de Cocultura , Vetores Genéticos , Antígenos de Histocompatibilidade Classe II/genética , Imunidade Celular/genética , Vírus da Coriomeningite Linfocítica/genética , Proteína 1 de Membrana Associada ao Lisossomo/genética , Proteína 1 de Membrana Associada ao Lisossomo/imunologia , Melanoma Experimental/imunologia , Melanoma Experimental/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Nucleocapsídeo/genética , Proteínas do Nucleocapsídeo/imunologia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/imunologia , Vacinas Virais/administração & dosagem , Vacinas Virais/genética
9.
Eur J Endocrinol ; 156(6): 663-71, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17535866

RESUMO

OBJECTIVE: The ATP-dependent K+-channel (K(ATP)) is critical for glucose sensing and normal glucagon and insulin secretion from pancreatic endocrine alpha- and beta-cells. Gastrointestinal endocrine L- and K-cells are also glucose-sensing cells secreting glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotrophic polypeptide (GIP) respectively. The aims of this study were to 1) investigate the expression and co-localisation of the K(ATP) channel subunits, Kir6.2 and SUR1, in human L- and K-cells and 2) investigate if a common hyperactive variant of the Kir6.2 subunit, Glu23Lys, exerts a functional impact on glucose-sensing tissues in vivo that may affect the overall glycaemic control in children with new-onset type 1 diabetes. DESIGN AND METHODS: Western blot and immunohistochemical analyses were performed for expression and co-localisation studies. Meal-stimulated C-peptide test was carried out in 257 children at 1, 6 and 12 months after diagnosis. Genotyping for the Glu23Lys variant was by PCR-restriction fragment length polymorphism. RESULTS: Kir6.2 and SUR1 co-localise with GLP-1 in L-cells and with GIP in K-cells in human ileum tissue. Children with type 1 diabetes carrying the hyperactive Glu23Lys variant had higher HbA1C at diagnosis (coefficient = 0.61%, P = 0.02) and 1 month after initial insulin therapy (coefficient = 0.30%, P = 0.05), but later disappeared. However, when adjusting HbA1C for the given dose of exogenous insulin, the dose-adjusted HbA1C remained higher throughout the 12 month study period (coefficient = 0.42%, P = 0.03). CONCLUSIONS: Kir6.2 and SUR1 co-localise in the gastrointestinal endocrine L- and K-cells. The hyperactive Glu23Lys variant of the K(ATP) channel subunit Kir6.2 may cause defective glucose sensing in several tissues and impaired glycaemic control in children with type 1 diabetes.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Canais de Potássio Corretores do Fluxo de Internalização/genética , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Canais de Potássio/metabolismo , Receptores de Droga/metabolismo , Adolescente , Western Blotting , Peptídeo C/metabolismo , Criança , Diabetes Mellitus Tipo 1/tratamento farmacológico , Ingestão de Alimentos/fisiologia , Feminino , Genótipo , Glucagon/metabolismo , Hemoglobinas Glicadas/metabolismo , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/genética , Hiperglicemia/metabolismo , Hipoglicemiantes/uso terapêutico , Íleo/citologia , Íleo/metabolismo , Imuno-Histoquímica , Insulina/uso terapêutico , Ilhotas Pancreáticas/metabolismo , Masculino , Polimorfismo de Fragmento de Restrição , Receptores de Sulfonilureias
10.
Am J Physiol Gastrointest Liver Physiol ; 293(1): G288-95, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17431213

RESUMO

Long-term treatment with dipeptidyl peptidase IV inhibitors (DPPIV-I) or glucagon-like peptide (GLP)-1 analogs may potentially affect intestinal growth by down- or upregulating the intestinotrophic hormone GLP-2. This study compared the intestinotrophic effects of 12-wk administration of vehicle, exendin-4 (Ex-4; 5 nmol/kg bid sc), or DPPIV-I (NN-7201, 10 mg/kg qd orally) in GK rats. Some animals were observed additionally for 9 wk after the end of treatment. Both treatments lowered glycated hemoglobin A1c at wk 12 vs. control (Ex-4, -0.8%; DPPIV-I, -0.4%). Body weight was reduced by Ex-4 compared with control (361 +/- 4 vs. 399 +/- 5 g; P < 0.001) because of reduced food intake, whereas neither parameter was affected by DPPIV-I. Linear bone growth was unaffected by either treatment. After treatment end, food intake in Ex-4 animals increased, and, by wk 21, body weight was identical in all groups. The small intestine of Ex-4-treated animals was larger at wk 12 compared with control (length, 135.6 +/- 1.6 vs. 124.5 +/- 2.3 cm, P < 0.001; absolute weight, 8.4 +/- 0.2 vs. 6.4 +/- 0.4 g, P < 0.001), being most pronounced proximally, where the absolute cross-sectional area related to body weight increased by 24% because of increased mucosal thickness. These effects were reversible, and 9 wk after the end of treatment, no differences between Ex-4 and control were apparent. Plasma GLP-2 concentrations were unaltered by either treatment, and Ex-4 had no agonistic or antagonistic effects on the transfected GLP-2 receptor. DPPIV-I had no intestinal effects. In conclusion, the continued presence of Ex-4 is necessary to maintain weight loss in GK rats. Effective antihyperglycemic treatment with Ex-4 increases intestinal mass reversibly, whereas DPPIV-I lacks intestinal effects.


Assuntos
Inibidores da Dipeptidil Peptidase IV , Intestino Delgado/patologia , Peptídeos/farmacologia , Inibidores de Proteases/farmacologia , Receptores de Glucagon/agonistas , Peçonhas/farmacologia , Xantinas/farmacologia , Animais , Peso Corporal , Células COS , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Tipo 2/patologia , Ingestão de Alimentos , Exenatida , Receptor do Peptídeo Semelhante ao Glucagon 1 , Receptor do Peptídeo Semelhante ao Glucagon 2 , Hemoglobinas Glicadas/metabolismo , Humanos , Intestino Delgado/efeitos dos fármacos , Masculino , Ratos , Receptores de Glucagon/efeitos dos fármacos , Transfecção
11.
J Biol Chem ; 281(16): 11050-7, 2006 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-16476726

RESUMO

The physiology of the incretin hormones, glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP), and their role in type 2 diabetes currently attract great interest. Recently we reported an essential role for prohormone convertase (PC) 1/3 in the cleavage of intestinal proglucagon, resulting in formation of GLP-1, as demonstrated in PC1/3-deficient mice. However, little is known about the endoproteolytic processing of the GIP precursor. This study investigates the processing of proGIP in PC1/3 and PC2 null mice and in cell lines using adenovirus-mediated overexpression. Supporting a role for PC1/3 in proGIP processing, we found co-localization of GIP and PC1/3 but not PC2 in intestinal sections by immunohistochemistry, and analysis of intestinal extracts from PC1/3-deficient animals demonstrated severely impaired processing to GIP, whereas processing to GIP was unaltered in PC2-deficient mice. Accordingly, overexpression of preproGIP in the neuroendocrine AtT-20 cell line that expresses high levels of endogenous PC1/3 and negligible levels of PC2 resulted in production of GIP. Similar results were obtained after co-expression of preproGIP and PC1/3 in GH4 cells that express no PC2 and only low levels of PC1/3. In addition, studies in GH4 cells and the alpha-TC1.9 cell line, expressing PC2 but not PC1/3, indicate that PC2 can mediate processing to GIP but also to other fragments not found in intestinal extracts. Taken together, our data indicate that PC1/3 is essential and sufficient for the production of the intestinal incretin hormone GIP, whereas PC2, although capable of cleaving proGIP, does not participate in intestinal proGIP processing and is not found in intestinal GIP-expressing cells.


Assuntos
Polipeptídeo Inibidor Gástrico/fisiologia , Glucose/metabolismo , Insulina/metabolismo , Pró-Proteína Convertase 1/fisiologia , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Células COS , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Cromatografia em Gel , Cromatografia Líquida de Alta Pressão , AMP Cíclico/metabolismo , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Citometria de Fluxo , Polipeptídeo Inibidor Gástrico/metabolismo , Vetores Genéticos , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Transgênicos , Modelos Genéticos , Peptídeos/química , Reação em Cadeia da Polimerase , Pró-Proteína Convertases/metabolismo , RNA/metabolismo , RNA Mensageiro/metabolismo , Radioimunoensaio , Ratos , Receptores dos Hormônios Gastrointestinais/metabolismo , Proteínas Recombinantes/química
12.
Blood ; 106(10): 3396-404, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16081685

RESUMO

Mitogen-activated protein kinase/extracellular signal to regulated kinase (MEK) kinase 1 (MEKK1) is a c-Jun N-terminal kinase (JNK) activating kinase known to be implicated in proinflammatory responses and cell motility. Using mice deficient for MEKK1 kinase activity (Mekk1(DeltaKD)) we show a role for MEKK1 in definitive mouse erythropoiesis. Although Mekk1(DeltaKD) mice are alive and fertile on a 129 x C57/BL6 background, the frequency of Mekk1(DeltaKD) embryos that develop past embryonic day (E) 14.5 is dramatically reduced when backcrossed into the C57/BL6 background. At E13.5, Mekk1(DeltaKD) embryos have normal morphology but are anemic due to failure of definitive erythropoiesis. When Mekk1(DeltaKD) fetal liver cells were transferred to lethally irradiated wild-type hosts, mature red blood cells were generated from the mutant cells, suggesting that MEKK1 functions in a non-cell-autonomous manner. Based on immunohistochemical and hemoglobin chain transcription analysis, we propose that the failure of definitive erythropoiesis is due to a deficiency in enucleation activity caused by insufficient macrophage-mediated nuclear DNA destruction.


Assuntos
Embrião de Mamíferos/embriologia , Eritropoese/genética , Hematopoese Extramedular/genética , Fígado/embriologia , MAP Quinase Quinase Quinase 1/metabolismo , Animais , Núcleo Celular/metabolismo , Núcleo Celular/patologia , DNA/genética , DNA/metabolismo , Embrião de Mamíferos/patologia , Hemoglobinas/genética , Hemoglobinas/metabolismo , Fígado/patologia , Transplante de Fígado/métodos , MAP Quinase Quinase Quinase 1/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Camundongos Knockout
13.
Regul Pept ; 124(1-3): 105-12, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15544847

RESUMO

BACKGROUND: Glucagon-like peptide-2 is thought to act as a growth factor for the gut, but the localization of the GLP-2 receptor and mechanism of action on epithelial growth is unclear. METHODS AND RESULTS: We found glucagon-like peptide-2 (GLP-2) receptors mainly on subepithelial myofibroblasts in rat, mouse, marmoset and human small and large intestine by immunohistochemistry and in situ hybridisation. By double labelling we found that these GLP-2 receptor immunoreactive cells also produce smooth muscle actin and keratinocyte growth factor (KGF). By subcutaneous infusion of either GLP-2 alone, GLP-2 plus KGF antibody, KGF antibody alone or saline in mice, we found that KGF antibody abolished the growth promoting effect of GLP-2 in the large intestine, but not in the small intestine. CONCLUSIONS: Our findings suggest that GLP-2 in the gut acts by activating receptors on the subepithelial myofibroblasts, causing the release of growth factors, which in turn stimulate intestinal growth.


Assuntos
Colo/efeitos dos fármacos , Colo/crescimento & desenvolvimento , Fatores de Crescimento de Fibroblastos/metabolismo , Peptídeos/farmacologia , Receptores de Glucagon/metabolismo , Actinas/metabolismo , Animais , Proliferação de Células , Colo/citologia , Colo/metabolismo , Feminino , Fator 7 de Crescimento de Fibroblastos , Fatores de Crescimento de Fibroblastos/antagonistas & inibidores , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Peptídeo 2 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Peptídeos Semelhantes ao Glucagon , Humanos , Camundongos , Ratos
14.
Diabetes ; 53 Suppl 3: S197-204, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15561911

RESUMO

Glucagon-like peptide (GLP)-1 is a gut hormone that stimulates insulin secretion, gene expression, and beta-cell growth. Together with the related hormone glucose-dependent insulinotropic polypeptide (GIP), it is responsible for the incretin effect, the augmentation of insulin secretion after oral as opposed to intravenous administration of glucose. Type 2 diabetic patients typically have little or no incretin-mediated augmentation of insulin secretion. This is due to decreased secretion of GLP-1 and loss of the insulinotropic effects of GIP. GLP-1, however, retains insulinotropic effects, and the hormone effectively improves metabolism in patients with type 2 diabetes. Continuous subcutaneous administration greatly improved glucose profiles and lowered body weight and HbA1c levels. Further, free fatty acid levels were lowered, insulin resistance was improved, and beta-cell performance was greatly improved. The natural peptide is rapidly degraded by the enzyme dipeptidyl peptidase IV (DPP IV), but resistant analogs as well as inhibitors of DPP IV are now under development, and both approaches have shown remarkable efficacy in experimental and clinical studies.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Hormônios Gastrointestinais/uso terapêutico , Glucagon/agonistas , Ilhotas Pancreáticas/metabolismo , Fragmentos de Peptídeos/agonistas , Precursores de Proteínas/agonistas , Animais , Modelos Animais de Doenças , Peptídeo 1 Semelhante ao Glucagon , Humanos
15.
Regul Pept ; 119(3): 151-4, 2004 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-15120474

RESUMO

BACKGROUND: The enteroinsular-axis is abnormal in type 2 diabetics, which contributes to the diabetic phenotype. The effect of the incretin hormone gastric inhibitory polypeptide (GIP) and the secretion of the incretin hormone glucagon-like peptide-1 (GLP-1) are thus greatly diminished. The explanation for these changes could be changes in the structure of either of the hormones or their receptors. Thus, the aim of this study was to study the occurrence of genetic variants in the GIP and GLP-1 encoding regions of the proGIP and proglucagon genes in type 2 diabetic patients and matched control subjects. METHODS AND RESULTS: Genomic DNA was extracted from buffy coats from 12 Caucasian type 2 diabetics and 12 healthy subjects, matched with respect to sex, age and BMI. The GIP and GLP-1 sequences were amplified using specific primers using the polymerase chain reaction (PCR). The amplified products were then sequenced. No germ-line mutations were identified in the GIP and the GLP-1 encoding regions of the proGIP and proglucagon genes in either the type 2 diabetic or the control subjects. CONCLUSIONS: The perturbed incretin effect in type 2 diabetics is not commonly caused by genetic variants in either the GIP or the GLP-1 encoding genes in type 2 diabetics.


Assuntos
Diabetes Mellitus Tipo 2/genética , Polipeptídeo Inibidor Gástrico/genética , Glucagon/genética , Fragmentos de Peptídeos/genética , Fenótipo , Precursores de Proteínas/genética , Idoso , Sequência de Bases , DNA/análise , Dinamarca , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Predisposição Genética para Doença , Glucagon/metabolismo , Peptídeo 1 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Humanos , Masculino , Pessoa de Meia-Idade , Dados de Sequência Molecular , Fragmentos de Peptídeos/metabolismo , Reação em Cadeia da Polimerase , Valor Preditivo dos Testes , Precursores de Proteínas/metabolismo , Receptores dos Hormônios Gastrointestinais/metabolismo , Receptores de Glucagon/metabolismo , Análise de Sequência de DNA , População Branca
16.
Endocrinology ; 145(3): 1349-55, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14630721

RESUMO

The neuroendocrine prohormone convertases 1 and 2 (PC1 and PC2) are expressed in endocrine intestinal L cells and pancreatic A cells, respectively, and colocalize with proglucagon in secretory granules. Mice lacking PC2 have multiple endocrinopathies and cannot process proglucagon to mature glucagon in the pancreas. Disruption of PC1 results in dwarfism and also multiple neuroendocrine peptide processing defects. This study compares the pancreatic and intestinal processing of proglucagon in mice lacking PC1 expression with that in age-matched wild-type controls. Because proglucagon was found to precipitate in acidic extracts, the intestinal processing profile was analyzed in both acidic and neutral extracts by gel filtration, HPLC, and RIA. Supporting a central role for PC2 in glucagon biosynthesis, we found normal processing of proglucagon to glucagon in the pancreas, whereas the intestinal proglucagon processing showed marked defects. Tissue proglucagon levels in null mice were elevated, and proglucagon processing to glicentin, oxyntomodulin, and glucagon-like peptide-1 and -2 (GLP-1 and GLP-2) was markedly decreased, indicating that PC1 is essential for the processing of all the intestinal proglucagon cleavage sites. This includes the monobasic site R(77) and, thereby, production of mature, biologically active GLP-1. We also found elevated glucagon levels, suggesting that factors other than PC1 that are capable of processing to mature glucagon are present in the secretory granules of the L cell. These findings strongly suggest that PC1 is essential for intestinal proglucagon processing in vivo and, thereby, plays an important role in production of the incretin hormone GLP-1 and the intestinotrophic hormone GLP-2.


Assuntos
Glucagon/metabolismo , Intestinos/enzimologia , Pró-Proteína Convertase 1/genética , Pró-Proteína Convertase 1/metabolismo , Precursores de Proteínas/metabolismo , Animais , Peptídeo 1 Semelhante ao Glucagon , Peptídeo 2 Semelhante ao Glucagon , Imuno-Histoquímica , Camundongos , Camundongos Mutantes , Pâncreas/enzimologia , Fragmentos de Peptídeos/metabolismo , Peptídeos/metabolismo , Proglucagon
17.
Regul Pept ; 114(2-3): 189-96, 2003 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12832109

RESUMO

BACKGROUND: The incretin hormones GIP and GLP-1 are thought to be produced in separate endocrine cells located in the proximal and distal ends of the mammalian small intestine, respectively. METHODS AND RESULTS: Using double immunohistochemistry and in situ hybridization, we found that GLP-1 was colocalized with either GIP or PYY in endocrine cells of the porcine, rat, and human small intestines, whereas GIP and PYY were rarely colocalized. Thus, of all the cells staining positively for either GLP-1, GIP, or both, 55-75% were GLP-1 and GIP double-stained in the mid-small intestine. Concentrations of extractable GIP and PYY were highest in the midjejunum [154 (95-167) and 141 (67-158) pmol/g, median and range, respectively], whereas GLP-1 concentrations were highest in the ileum [92 (80-207) pmol/l], but GLP-1, GIP, and PYY immunoreactive cells were found throughout the porcine small intestine. CONCLUSIONS: Our results provide a morphological basis to suggest simultaneous, rather than sequential, secretion of these hormones by postprandial luminal stimulation.


Assuntos
Glândulas Endócrinas/metabolismo , Polipeptídeo Inibidor Gástrico/metabolismo , Glucagon/metabolismo , Intestino Delgado/metabolismo , Fragmentos de Peptídeos/metabolismo , Precursores de Proteínas/metabolismo , Animais , Cromatografia em Gel , Glândulas Endócrinas/citologia , Feminino , Peptídeo 1 Semelhante ao Glucagon , Hibridização In Situ , Intestino Delgado/citologia , Suínos
18.
Regul Pept ; 105(3): 173-9, 2002 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-11959371

RESUMO

Supraphysiological doses of glucagon-like peptide-2 (GLP-2) have been shown to induce intestinal growth by increasing villus height and crypt depth and by decreasing apoptosis, but a physiological effect of GLP-2 has not yet been demonstrated. Earlier, we found elevated levels of endogenous GLP-2 in untreated streptozotocin diabetic rats associated with marked intestinal growth. In the present study, we investigated the role of endogenous GLP-2 for this adaptive response. We included four groups of six rats: (1) diabetic rats treated with saline, (2) diabetic rats treated with non-specific antibodies, (3) diabetic rats treated with polyclonal GLP-2 antibodies and (4) non-diabetic control rats treated with saline. All animals were treated with once daily intraperitoneal injections for 13 days and killed on day 14. Diabetic rats treated with saline or non-specific antibodies had a significantly (P<0.01) increased area of mucosa (13.00+/-0.64 and 13.37+/-0.60 mm(2), respectively) in the proximal part of the small intestine compared with non-diabetic controls (7.97+/-0.70 mm(2)). In contrast, diabetic rats treated with GLP-2 antibodies had a significantly (P<0.01) smaller increase in area of mucosa in the proximal part of the small intestine (10.84+/-0.44 mm(2)). Antibody treatment had no effect on body weight, blood glucose concentrations and food intake. Thus, blocking of endogenous GLP-2 in a model of adaptive intestinal growth reduces the growth response, providing strong evidence for a physiological growth factor function of GLP-2.


Assuntos
Diabetes Mellitus/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/crescimento & desenvolvimento , Peptídeos/antagonistas & inibidores , Peptídeos/metabolismo , Animais , Glicemia , Peso Corporal , Ingestão de Alimentos , Feminino , Peptídeo 2 Semelhante ao Glucagon , Peptídeos Semelhantes ao Glucagon , Intestinos/anatomia & histologia , Testes de Neutralização , Tamanho do Órgão , Peptídeos/sangue , Peptídeos/imunologia , Ratos , Ratos Wistar
19.
Regul Pept ; 103(1): 9-15, 2002 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-11738243

RESUMO

The therapeutic potential of the intestinotrophic mediator glucagon-like peptide-2 (1-33) [GLP-2 (1-33)] has increased interest in the pharmacokinetics of the peptide. This study was undertaken to investigate whether the primary degradation product GLP-2 (3-33) interacts with the GLP-2 receptor. Functional (cAMP) and binding in vitro studies were carried out in cells expressing the transfected human GLP-2 receptor. Furthermore, a biologic response of GLP-2 (3-33) was tested in vivo. Mice were allocated to groups treated for 10 days (twice daily) with: (1) 5 microg GLP-2 (1-33), (2) 25 microg GLP-2 (3-33), (3) 5 microg GLP-2 (1-33)+100 microg GLP-2 (3-33), or (4) 5 microg GLP-2 (1-33)+500 microg GLP-2 (3-33). The intestine was investigated for growth changes. GLP-2 (3-33) bound to the GLP-2 receptor with a binding affinity of 7.5% of that of GLP-2 (1-33). cAMP accumulation was stimulated with an efficacy of 15% and a potency more than two orders of magnitude lower than that of GLP-2 (1-33). Increasing doses of GLP-2 (3-33) (10(-7)-10(-5) M) caused a shift to the right in the dose-response curve of GLP-2 (1-33). Treatment of mice with either GLP-2 (1-33) or (3-33) induced significant growth responses in both the small and large intestines, but the response induced by GLP-2 (3-33) was much smaller. Co-administration of 500 microg of GLP-2 (3-33) and 5 microg GLP-2 (1-33) resulted in a growth response that was smaller than that of 5 microg GLP-2 (1-33) alone. Consistent with the observed in vivo activities, our functional studies and binding data indicate that GLP-2 (3-33) acts as a partial agonist with potential competitive antagonistic properties on the GLP-2 receptor.


Assuntos
Intestino Grosso/crescimento & desenvolvimento , Intestino Delgado/crescimento & desenvolvimento , Fragmentos de Peptídeos/farmacologia , Receptores de Glucagon/agonistas , Animais , Peso Corporal , Linhagem Celular , Cricetinae , AMP Cíclico/metabolismo , Esquema de Medicação , Feminino , Peptídeo 2 Semelhante ao Glucagon , Receptor do Peptídeo Semelhante ao Glucagon 1 , Peptídeos Semelhantes ao Glucagon , Humanos , Processamento de Imagem Assistida por Computador , Injeções Subcutâneas , Intestino Grosso/citologia , Intestino Grosso/efeitos dos fármacos , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão/efeitos dos fármacos , Fragmentos de Peptídeos/química , Ligação Proteica , Distribuição Aleatória , Receptores de Glucagon/metabolismo , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA