Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Mol Metab ; 66: 101638, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36400403

RESUMEN

OBJECTIVE: Glucose-dependent insulinotropic polypeptide (GIP) is one of the two major incretin factors that regulate metabolic homeostasis. Genetic ablation of its receptor (GIPR) in mice confers protection against diet-induced obesity (DIO), while GIPR neutralizing antibodies produce additive weight reduction when combined with GLP-1R agonists in preclinical models and clinical trials. Conversely, GIPR agonists have been shown to promote weight loss in rodents, while dual GLP-1R/GIPR agonists have proven superior to GLP-1R monoagonists for weight reduction in clinical trials. We sought to develop a long-acting, specific GIPR peptide antagonist as a tool compound suitable for investigating GIPR pharmacology in both rodent and human systems. METHODS: We report a structure-activity relationship of GIPR peptide antagonists based on the human and mouse GIP sequences with fatty acid-based protraction. We assessed these compounds in vitro, in vivo in DIO mice, and ex vivo in islets from human donors. RESULTS: We report the discovery of a GIP(5-31) palmitoylated analogue, [Nα-Ac, L14, R18, E21] hGIP(5-31)-K11 (γE-C16), which potently inhibits in vitro GIP-mediated cAMP generation at both the hGIPR and mGIPR. In vivo, this peptide effectively blocks GIP-mediated reductions in glycemia in response to exogenous and endogenous GIP and displays a circulating pharmacokinetic profile amenable for once-daily dosing in rodents. Co-administration with the GLP-1R agonist semaglutide and this GIPR peptide antagonist potentiates weight loss compared to semaglutide alone. Finally, this antagonist inhibits GIP- but not GLP-1-stimulated insulin secretion in intact human islets. CONCLUSIONS: Our work demonstrates the discovery of a potent, specific, and long-acting GIPR peptide antagonist that effectively blocks GIP action in vitro, ex vivo in human islets, and in vivo in mice while producing additive weight-loss when combined with a GLP-1R agonist in DIO mice.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón , Receptores de la Hormona Gastrointestinal , Roedores , Animales , Humanos , Ratones , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Ratones Obesos , Péptidos/farmacología , Péptidos/química , Roedores/metabolismo , Pérdida de Peso , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores
2.
Endocrinology ; 162(7)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33782700

RESUMEN

The incretin effect-the amplification of insulin secretion after oral vs intravenous administration of glucose as a mean to improve glucose tolerance-was suspected even before insulin was discovered, and today we know that the effect is due to the secretion of 2 insulinotropic peptides, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). But how important is it? Physiological experiments have shown that, because of the incretin effect, we can ingest increasing amounts of amounts of glucose (carbohydrates) without increasing postprandial glucose excursions, which otherwise might have severe consequences. The mechanism behind this is incretin-stimulated insulin secretion. The availability of antagonists for GLP-1 and most recently also for GIP has made it possible to directly estimate the individual contributions to postprandial insulin secretion of a) glucose itself: 26%; b) GIP: 45%; and c) GLP-1: 29%. Thus, in healthy individuals, GIP is the champion. When the action of both incretins is prevented, glucose tolerance is pathologically impaired. Thus, after 100 years of research, we now know that insulinotropic hormones from the gut are indispensable for normal glucose tolerance. The loss of the incretin effect in type 2 diabetes, therefore, contributes greatly to the impaired postprandial glucose control.


Asunto(s)
Glucemia/fisiología , Polipéptido Inhibidor Gástrico/fisiología , Péptido 1 Similar al Glucagón/fisiología , Homeostasis/fisiología , Incretinas/fisiología , Insulina/fisiología , Diabetes Mellitus Tipo 2/fisiopatología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Humanos , Secreción de Insulina/efectos de los fármacos , Periodo Posprandial , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores
3.
Int J Mol Sci ; 21(4)2020 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-32098413

RESUMEN

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are gut hormones that are secreted from enteroendocrine L cells and K cells in response to digested nutrients, respectively. They are also referred to incretin for their ability to stimulate insulin secretion from pancreatic beta cells in a glucose-dependent manner. Furthermore, GLP-1 exerts anorexic effects via its actions in the central nervous system. Since native incretin is rapidly inactivated by dipeptidyl peptidase-4 (DPP-4), DPP-resistant GLP-1 receptor agonists (GLP-1RAs), and DPP-4 inhibitors are currently used for the treatment of type 2 diabetes as incretin-based therapy. These new-class agents have superiority to classical oral hypoglycemic agents such as sulfonylureas because of their low risks for hypoglycemia and body weight gain. In addition, a number of preclinical studies have shown the cardioprotective properties of incretin-based therapy, whose findings are further supported by several randomized clinical trials. Indeed, GLP-1RA has been significantly shown to reduce the risk of cardiovascular and renal events in patients with type 2 diabetes. However, the role of GIP in cardiovascular disease remains to be elucidated. Recently, pharmacological doses of GIP receptor agonists (GIPRAs) have been found to exert anti-obesity effects in animal models. These observations suggest that combination therapy of GLP-1R and GIPR may induce superior metabolic and anti-diabetic effects compared with each agonist individually. Clinical trials with GLP-1R/GIPR dual agonists are ongoing in diabetic patients. Therefore, in this review, we summarize the cardiovascular effects of GIP and GIPRAs in cell culture systems, animal models, and humans.


Asunto(s)
Aterosclerosis/metabolismo , Enfermedades Cardiovasculares/metabolismo , Células Enteroendocrinas/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Animales , Aterosclerosis/tratamiento farmacológico , Glucemia/metabolismo , Enfermedades Cardiovasculares/tratamiento farmacológico , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Células Enteroendocrinas/citología , Células Enteroendocrinas/efectos de los fármacos , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Humanos , Secreción de Insulina/efectos de los fármacos
4.
Peptides ; 125: 170203, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31733230

RESUMEN

Gastric inhibitory polypeptide (GIP) is a 42 amino acid hormone secreted from intestinal K-cells in response to nutrient ingestion. Despite a recognised physiological role for GIP as an insulin secretagogue to control postprandial blood glucose levels, growing evidence reveals important actions of GIP on adipocytes and promotion of fat deposition in tissues. As such, blockade of GIP receptor (GIPR) action has been proposed as a means to counter insulin resistance, and improve metabolic status in obesity and related diabetes. In agreement with this, numerous independent observations in animal models support important therapeutic applications of GIPR antagonists in obesity-diabetes. Sustained administration of peptide-based GIPR inhibitors, low molecular weight GIPR antagonists, GIPR neutralising antibodies as well as genetic knockout of GIPR's or vaccination against GIP all demonstrate amelioration of insulin resistance and reduced body weight gain in response to high fat feeding. These observations were consistently associated with decreased accumulation of lipids in peripheral tissues, thereby alleviating insulin resistance. Although the impact of prolonged GIPR inhibition on bone turnover still needs to be determined, evidence to date indicates that GIPR antagonists represent an exciting novel treatment option for obesity-diabetes.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Resistencia a la Insulina , Insulina/metabolismo , Obesidad/fisiopatología , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Pérdida de Peso/efectos de los fármacos , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Humanos
5.
Peptides ; 125: 170227, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31805296

RESUMEN

Gastric inhibitory polypeptide (GIP) is a regulatory peptide expressed in the mammalian upper small intestine, and both GIP and its receptor (GIPR) are expressed in the cortex and hippocampus regions of the brain as well. While learning and memory deficits have been observed in GIPR-/- mice, the effects of peripheral GIP immunoneutralization on motor-coordination, learning, and memory have not been examined. In the present study, adult GIPR-/- mice (KO) and age-matched wild-type C57BL/6 J mice (WT) received weekly vehicle PBS injections for 12 weeks, while a third group of wild-type mice were injected weekly for 12 weeks with 30 mg/kg body weight humanized GIP-mAb (AB) to assess the possibility of long-term effects of peripheral GIP antagonism on rodent memory and behavior. All mice groups then underwent a battery of tests that evaluated motor behavior, body coordination, and memory. Performance deficits in several memory studies after 12 weeks of treatment were demonstrated in KO, but not in AB or WT mice. Body coordination performance showed no significant differences among the 3 groups. A similar short-term study (3 injections over 9 days) was also conducted and the results were similar to those from the long-term study. Thus, short-term and long-term peripheral GIP antagonism by GIP-mAb did not appear to affect learning and memory in mice, consistent with the notion that the GIP-mAb does not cross the blood brain barrier. Furthermore, our studies indicate that GIP signaling in the brain appears to involve local neurocrine pathways.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Aprendizaje/efectos de los fármacos , Memoria/efectos de los fármacos , Actividad Motora/efectos de los fármacos , Receptores de la Hormona Gastrointestinal/fisiología , Animales , Modelos Animales de Enfermedad , Polipéptido Inhibidor Gástrico/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
6.
Biochem Biophys Res Commun ; 513(4): 974-982, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31003779

RESUMEN

AIMS/INTRODUCTION: Caloric restriction (CR) promotes longevity and exerts anti-aging effects by increasing Sirtuin production and activation. Gastric inhibitory polypeptide (GIP), a gastrointestinal peptide hormone, exerts various effects on pancreatic ß-cells and extra-pancreatic tissues. GIP promotes glucose-dependent augmentation of insulin secretion and uptake of nutrients into the adipose tissue. MATERIALS AND METHODS: Gipr-/- and Gipr+/+ mice were used for lifespan analysis, behavior experiments and gene expression of adipose tissue and muscles. 3T3-L1 differentiated adipocytes were used for Sirt1 and Nampt expression followed by treatment with GIP and α-lipoic acid. RESULTS: We observed that GIP receptor-knockout (Gipr-/-) mice fed normal diet showed an extended lifespan, increased exploratory and decreased anxiety-based behaviors, which are characteristic behavioral changes under CR. Moreover, Gipr-/- mice showed increased Sirt1 and Nampt expression in the adipose tissue. GIP suppressed α-lipoic acid-induced Sirt1 expression and activity in differentiated adipocytes. CONCLUSIONS: Although maintenance of CR is difficult, food intake and muscle endurance of Gipr-/- mice were similar to those of wild-type mice. Inhibition of GIP signaling may be a novel strategy to extend the lifespan of diabetic patients.


Asunto(s)
Restricción Calórica , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Longevidad/fisiología , Transducción de Señal/fisiología , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Animales , Citocinas/metabolismo , Ratones , Ratones Noqueados , Nicotinamida Fosforribosiltransferasa/metabolismo , Receptores de la Hormona Gastrointestinal/genética , Sirtuina 1/metabolismo
7.
Peptides ; 100: 190-201, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29412819

RESUMEN

Chemical derivatives of the gut-derived peptide hormone glucagon-like peptide 1 (GLP-1) are among the best-in-class pharmacotherapies to treat obesity and type 2 diabetes. However, GLP-1 analogs have modest weight lowering capacity, in the range of 5-10%, and the therapeutic window is hampered by dose-dependent side effects. Over the last few years, a new concept has emerged: combining the beneficial effects of several key metabolic hormones into a single molecular entity. Several unimolecular GLP-1-based polyagonists have shown superior metabolic action compared to GLP-1 monotherapies. In this review article, we highlight the history of polyagonists targeting the receptors for GLP-1, GIP and glucagon, and discuss recent progress in expanding of this concept to now allow targeted delivery of nuclear hormones via GLP-1 and other gut hormones, as a novel approach towards more personalized pharmacotherapies.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/uso terapéutico , Péptido 1 Similar al Glucagón/uso terapéutico , Obesidad/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Hormonas Gastrointestinales/antagonistas & inhibidores , Hormonas Gastrointestinales/genética , Hormonas Gastrointestinales/uso terapéutico , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor del Péptido 1 Similar al Glucagón/genética , Humanos , Incretinas/metabolismo , Insulina/genética , Insulina/metabolismo , Obesidad/metabolismo , Obesidad/patología , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Receptores de la Hormona Gastrointestinal/genética , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/genética
8.
Br J Pharmacol ; 173(5): 888-98, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26661062

RESUMEN

BACKGROUND AND PURPOSE: Galanin is a widely expressed neuropeptide, which in the gut is thought to modulate gastrointestinal motility and secretion. We aimed to elucidate the poorly characterised mechanisms underlying the inhibitory effect of galanin and the potential involvement of G-protein coupled inwardly rectifying potassium, Kir 3, (GIRK) channels in glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) secretion. EXPERIMENTAL APPROACH: Purified murine L and K cells were analysed for expression of galanin receptors and GIRK subunits. Hormone secretion was measured from primary murine intestinal cultures. Intracellular cAMP was monitored in primary L cells derived from mice expressing the Epac2camps sensor under the control of the proglucagon promoter. KEY RESULTS: Galanin receptor 1 (GAL1, Galr1) and GIRK channel 1 (Kir 3.1, Kcnj3) and 4 (Kir 3.4, Kcnj5) mRNA expression was highly enriched in K and L cells. Galanin and a selective GAL1 receptor agonist (M617) potently inhibited GLP-1 and GIP secretion from primary small intestinal cultures. In L cells, galanin significantly inhibited the forskolin-induced cAMP response. The GIRK1/4 activator ML297 significantly reduced glucose-stimulated and IBMX-stimulated GLP-1 secretion but had no effect on GIP. The GIRK blocker tertiapin-Q did not impair galanin-mediated GLP-1 inhibition. CONCLUSIONS AND IMPLICATIONS: Galanin, acting via the GAL1 receptor and Gi -coupled signalling in L and K cells, is a potent inhibitor of GLP-1 and GIP secretion. Although GIRK1/4 channels are expressed in these cells, their activation does not appear to play a major role in galanin-mediated inhibition of incretin secretion.


Asunto(s)
Células Enteroendocrinas/efectos de los fármacos , Galanina/farmacología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Receptor de Galanina Tipo 1/genética , Animales , Células Cultivadas , AMP Cíclico/metabolismo , Células Enteroendocrinas/metabolismo , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , ARN Mensajero/metabolismo
9.
Am J Physiol Endocrinol Metab ; 309(12): E1008-18, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26487006

RESUMEN

Previous reports have suggested that the abrogation of gastric inhibitory polypeptide (GIP) signaling could be exploited to prevent and treat obesity and obesity-related disorders in humans. This study was designed to determine whether immunoneutralization of GIP, using a newly developed specific monoclonal antibody (mAb), would prevent the development of obesity. Specific mAb directed against the carboxy terminus of mouse GIP was identified, and its effects on the insulin response to oral and to intraperitoneal (ip) glucose and on weight gain were evaluated. Administration of mAb (30 mg/kg body wt, BW) to mice attenuated the insulin response to oral glucose by 70% and completely eliminated the response to ip glucose coadministered with human GIP. Nine-week-old C57BL/6 mice injected with GIP mAbs (60 mg·kg BW(-1)·wk(-1)) for 17 wk gained 46.5% less weight than control mice fed an identical high-fat diet (P < 0.001). No significant differences in the quantity of food consumed were detected between the two treatment groups. Furthermore, magnetic resonance imaging demonstrated that subcutaneous, omental, and hepatic fat were 1.97-, 3.46-, and 2.15-fold, respectively, lower in mAb-treated animals than in controls. Moreover, serum insulin, leptin, total cholesterol (TC), low-density lipoprotein (LDL), and triglycerides were significantly reduced, whereas the high-density lipoprotein (HDL)/TC ratio was 1.25-fold higher in treated animals than in controls. These studies support the hypothesis that a reduction in GIP signaling using a GIP-neutralizing mAb might provide a useful method for the treatment and prevention of obesity and related disorders.


Asunto(s)
Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/inmunología , Polipéptido Inhibidor Gástrico/inmunología , Obesidad/inmunología , Obesidad/prevención & control , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Inmunoterapia/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Obesidad/diagnóstico , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología , Resultado del Tratamiento
10.
Biomed Khim ; 61(4): 488-96, 2015.
Artículo en Ruso | MEDLINE | ID: mdl-26350740

RESUMEN

This review summarizes data on the main approaches used for the search of biologically active compounds modulating the level and physiological activity of incretins. Currently two groups of drugs are used in clinical practice: they either replenish the deficit of incretins (glucagon-like peptide-1 receptor agonists) or inhibit the degradation processes (dipeptidyl peptidase 4 inhibitors). In addition, new groups of substances are actively searched. These include non-peptide agonists of glucagon-like peptide-1 receptors, agonists/antagonists of glucose-dependent insulinotropic peptide, the hybrid polypeptides based on glucagon-like peptide-1 and glucagon.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Hipoglucemiantes/uso terapéutico , Peptidomiméticos/uso terapéutico , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Secuencia de Aminoácidos , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Descubrimiento de Drogas , Polipéptido Inhibidor Gástrico/agonistas , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/genética , Polipéptido Inhibidor Gástrico/metabolismo , Regulación de la Expresión Génica , Glucagón/química , Glucagón/metabolismo , Péptido 1 Similar al Glucagón/genética , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Humanos , Hipoglucemiantes/química , Datos de Secuencia Molecular , Peptidomiméticos/química , Transducción de Señal , Bibliotecas de Moléculas Pequeñas/química
11.
J Diabetes Res ; 2015: 326359, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26221611

RESUMEN

Glucose-dependent insulinotropic peptide (GIP) is an incretin hormone produced in the gastrointestinal tract that stimulates glucose dependent insulin secretion. Impaired incretin response has been documented in diabetic patients and was mainly related to the inability of the pancreatic beta cells to secrete insulin in response to GIP. Advanced Glycation End Products (AGEs) have been shown to play an important role in pancreatic beta cell dysfunction. The aim of this study is to investigate whether the exposure to AGEs can induce GIP resistance in the pancreatic beta cell line HIT-T15. Cells were cultured for 5 days in low (CTR) or high glucose (HG) concentration in the presence of AGEs (GS) to evaluate the expression of GIP receptor (GIPR), the intracellular signaling activated by GIP, and secretion of insulin in response to GIP. The results showed that incubation with GS alone altered intracellular GIP signaling and decreased insulin secretion as compared to CTR. GS in combination with HG reduced the expression of GIPR and PI3K and abrogated GIP-induced AKT phosphorylation and GIP-stimulated insulin secretion. In conclusion, we showed that treatment with GS is associated with the loss of the insulinotropic effect of GIP in hyperglycemic conditions.


Asunto(s)
Regulación hacia Abajo , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Productos Finales de Glicación Avanzada/metabolismo , Hiperglucemia/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Animales , Glucemia/análisis , Western Blotting , Línea Celular , Polipéptido Inhibidor Gástrico/sangre , Polipéptido Inhibidor Gástrico/genética , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/metabolismo , Productos Finales de Glicación Avanzada/sangre , Humanos , Hiperglucemia/sangre , Secreción de Insulina , Mesocricetus , Fosfatidilinositol 3-Quinasa/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de la Hormona Gastrointestinal/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transducción de Señal
12.
Diabetologia ; 58(9): 2144-53, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26048235

RESUMEN

AIMS/HYPOTHESIS: GIP(6-30)Cex-K(40)[Pal] has been characterised as a fatty-acid-derived gastric inhibitory polypeptide (GIP) inhibitor that can induce pancreatic beta cell rest by diminishing the incretin effect. We investigated its therapeutic efficacy with and without the glucagon-like peptide-1 (GLP-1) beta cell cytotropic agent liraglutide. METHODS: The therapeutic efficacy of GIP(6-30)Cex-K(40)[Pal] alone, and in combination with liraglutide, was determined in C57BL/KsJ db/db mice using a sequential 12 h administration schedule. RESULTS: GIP(6-30)Cex-K(40)[Pal] was devoid of cAMP-generating or insulin-secretory activity, and inhibited GIP-induced cAMP production and insulin secretion. GIP(6-30)Cex-K(40)[Pal] also inhibited GIP-induced glucose-lowering and insulin-releasing actions in mice. Dose- and time-dependent studies in mice revealed that 2.5 nmol/kg GIP(6-30)Cex-K(40)[Pal], and 0.25 nmol/kg liraglutide, imparted distinct biological effects for 8-12 h post administration. When GIP(6-30)Cex-K(40)[Pal] (2.5 nmol/kg) and liraglutide (0.25 nmol/kg) were administered sequentially at 12 h intervals (at 08:00 and 20:00 hours) to db/db mice for 28 days, mice treated with GIP(6-30)Cex-K(40)[Pal] (08:00 hours) and liraglutide (20:00 hours) displayed pronounced reductions in circulating glucose and insulin. Both oral and intraperitoneal glucose tolerance and glucose-stimulated plasma insulin concentrations were improved together with enhanced insulin sensitivity. The expression of genes involved in adipocyte lipid deposition was generally decreased. The other treatment modalities, including GIP(6-30)Cex-K(40)[Pal] (08:00 and 20:00 hours), liraglutide (08:00 and 20:00 hours) and liraglutide (08:00 hours) combined with GIP(6-30)Cex-K(40)[Pal] (20:00 hours), also imparted beneficial effects but these were not as prominent as those of GIP(6-30)Cex-K(40)[Pal] (08:00 hours) and liraglutide (20:00 hours). CONCLUSION/INTERPRETATION: These data demonstrate that periods of beta cell rest combined with intervals of beta cell stimulation benefit diabetes control and should be further evaluated as a potential treatment option for type 2 diabetes.


Asunto(s)
Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Células Secretoras de Insulina/citología , Administración Oral , Animales , Glucemia/análisis , Cricetinae , AMP Cíclico/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Esquema de Medicación , Ácidos Grasos/metabolismo , Homeostasis , Humanos , Incretinas/metabolismo , Infusiones Parenterales , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Liraglutida/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
13.
Mol Cell Endocrinol ; 401: 120-9, 2015 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-25449420

RESUMEN

Compromise of gastric inhibitory polypeptide (GIP) receptor signalling represents a possible therapeutic strategy for the treatment of obesity-related diabetes. This study has characterised and evaluated the C-terminally fatty acid derivatised GIP analogues, GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal], as potential GIP inhibitors. Both GIP analogues lack the two N-terminal amino acids cleaved by DPP-4 and have addition of nine amino acids from the C-terminal of exendin(1-39), Cex. GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] effectively (p < 0.01 to p < 0.001) inhibited GIP-induced cAMP production and insulin secretion in vitro. In normal mice, GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] displayed a significant (p < 0.05 to p < 0.001) and prolonged inhibitory effect on GIP-induced glucose-lowering and insulin-releasing actions. When injected once daily for 21 days in obese-diabetic high fat fed mice, both GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] significantly reduced body weight (p < 0.01 to p < 0.001) and lowered circulating glucose (p < 0.001) and insulin (p < 0.01 to p < 0.001) concentrations. The observed beneficial changes were independent of effects on energy intake, locomotor activity or metabolic rate. Oral and intraperitoneal glucose tolerance were significantly (p < 0.05 to p < 0.001) improved in both treatment groups at the end of the study, despite reduced glucose-induced plasma insulin concentrations. This improvement of metabolic control was accompanied by enhanced (p < 0.05 to p < 0.01) insulin sensitivity compared with high fat controls. These data demonstrate the potential offered by GIP(3-30)Cex-K(40)[Pal] and Pro(3)GIP(3-30)Cex-K(40)[Pal] for the treatment of obesity-related diabetes.


Asunto(s)
Fármacos Antiobesidad/administración & dosificación , Dieta Alta en Grasa/efectos adversos , Polipéptido Inhibidor Gástrico/análogos & derivados , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Administración Oral , Animales , Glucemia/metabolismo , Peso Corporal/efectos de los fármacos , Esquema de Medicación , Polipéptido Inhibidor Gástrico/química , Prueba de Tolerancia a la Glucosa , Inyecciones Intraperitoneales , Insulina/sangre , Lipoilación , Ratones , Obesidad/inducido químicamente
14.
Nutr Res ; 34(8): 653-60, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25172378

RESUMEN

Insulin resistance is central to the etiology of the metabolic syndrome cluster of diseases. Evidence suggests that a high-fat diet is associated with insulin resistance, which may be modulated by dietary fatty acid composition. We hypothesized that high saturated fatty acid intake increases insulin and gastric inhibitory polypeptide (GIP) secretion. To clarify the effect of ingested fatty acid composition on glucose levels, we conducted an intervention study to investigate the insulin and plasma GIP responses in 11 healthy women, including a dietary control. Subjects were provided daily control meals (F-20; saturated fatty acids/monounsaturated fatty acids/polyunsaturated fatty acids [S/M/P] ratio, 3:4:3) with 20 energy (E) % fat, followed by 2 isoenergetic experimental meals for 7 days each. These meals comprised 60 E% carbohydrate, 15 E% protein, and 30 E% fat (FB-30; high saturated fatty acid meal; S/M/P, 5:4:1; F-30: reduced saturated fatty acid meal; S/M/P, 3:4:3). On the second day of the F-20 and the last day of F-30 and FB-30, blood samples were taken before and 30, 60, and 120 minutes after a meal tolerance test. The plasma glucose responses did not differ between F-20 and FB-30 or F-30. However, insulin levels were higher after the FB-30 than after the F-20 (P < .01). The GIP response after the FB-30 was higher than that after the F-30 (P < .05). In addition, the difference in the incremental GIP between FB-30 and F-30 correlated significantly and positively with that of the insulin. These results suggest that a high saturated fatty acid content stimulates postprandial insulin release via increased GIP secretion.


Asunto(s)
Glucemia/metabolismo , Dieta Alta en Grasa , Grasas de la Dieta/farmacología , Ácidos Grasos/farmacología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Insulina/sangre , Adulto , Ácidos Grasos/administración & dosificación , Femenino , Humanos , Valores de Referencia , Adulto Joven
15.
Endocrinology ; 154(9): 3089-98, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23798598

RESUMEN

We have previously demonstrated that ileal administration of the dietary protein hydrolysate prepared from corn zein (ZeinH) stimulated glucagon-like peptide-1 (GLP-1) secretion and attenuated hyperglycemia in rats. In this study, to examine whether oral administration of ZeinH improves glucose tolerance by stimulating GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) secretion, glucose tolerance tests were performed in normal Sprague-Dawley male rats and diabetic Goto-Kakizaki (GK) male rats. The test solution was gavaged before ip glucose injection in normal rats or gavaged together with glucose in GK rats. Blood samples were collected from the tail vein or by using the jugular catheter to measure glucose, insulin, GLP-1, and GIP levels. In the ip glucose tolerance test, oral administration of ZeinH (2 g/kg) significantly suppressed the glycemic response accompanied by an immediate increase in plasma GLP-1 and GIP levels in normal rats. In contrast, oral administration of another dietary peptide, meat hydrolysate, did not elicit a similar effect. The glucose-lowering effect of ZeinH was attenuated by a GLP-1 receptor antagonist or by a GIP receptor antagonist. Furthermore, oral ZeinH induced GLP-1 secretion and reduced glycemic response in GK rats under the oral glucose tolerance test. These results indicate that the oral administration of the dietary peptide ZeinH improves glucose tolerance in normal and diabetic rats by its incretin-releasing activity, namely, the incretinotropic effect.


Asunto(s)
Diabetes Mellitus Tipo 2/dietoterapia , Enterocitos/metabolismo , Polipéptido Inhibidor Gástrico/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Hipoglucemiantes/uso terapéutico , Hidrolisados de Proteína/uso terapéutico , Zeína/uso terapéutico , Animales , Línea Celular , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/metabolismo , Suplementos Dietéticos , Digestión , Enterocitos/efectos de los fármacos , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/sangre , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/sangre , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Hipoglucemiantes/administración & dosificación , Hipoglucemiantes/antagonistas & inhibidores , Hipoglucemiantes/metabolismo , Masculino , Ratones , Hidrolisados de Proteína/administración & dosificación , Hidrolisados de Proteína/antagonistas & inhibidores , Hidrolisados de Proteína/metabolismo , Ratas , Ratas Endogámicas , Ratas Sprague-Dawley , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Zeína/administración & dosificación , Zeína/antagonistas & inhibidores , Zeína/metabolismo
16.
Diabetes Obes Metab ; 14(2): 101-11, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21752172

RESUMEN

Type 2 diabetes mellitus (T2DM) develops as a consequence of progressive ß-cell dysfunction in the presence of insulin resistance. None of the currently-available T2DM therapies is able to change the course of the disease by halting the relentless decline in pancreatic islet cell function. Recently, dipeptidyl peptidase (DPP)-4 inhibitors, or incretin enhancers, have been introduced in the treatment of T2DM. This class of glucose-lowering agents enhances endogenous glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) levels by blocking the incretin-degrading enzyme DPP-4. DPP-4 inhibitors may restore the deranged islet-cell balance in T2DM, by stimulating meal-related insulin secretion and by decreasing postprandial glucagon levels. Moreover, in rodent studies, DPP-4 inhibitors demonstrated beneficial effects on (functional) ß-cell mass and pancreatic insulin content. Studies in humans with T2DM have indicated improvement of islet-cell function, both in the fasted state and under postprandial conditions and these beneficial effects were sustained in studies with a duration up to 2 years. However, there is at present no evidence in humans to suggest that DPP-4 inhibitors have durable effects on ß-cell function after cessation of therapy. Long-term, large-sized trials using an active blood glucose lowering comparator followed by a sufficiently long washout period after discontinuation of the study drug are needed to assess whether DPP-4 inhibitors may durably preserve pancreatic islet-cell function in patients with T2DM.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Dipeptidil Peptidasa 4/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Péptido 1 Similar al Glucagón/antagonistas & inhibidores , Islotes Pancreáticos/efectos de los fármacos , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/sangre , Progresión de la Enfermedad , Perros , Polipéptido Inhibidor Gástrico/sangre , Polipéptido Inhibidor Gástrico/efectos de los fármacos , Péptido 1 Similar al Glucagón/sangre , Péptido 1 Similar al Glucagón/efectos de los fármacos , Humanos , Ratas
17.
Best Pract Res Clin Endocrinol Metab ; 23(4): 499-512, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19748067

RESUMEN

Glucose-dependent insulinotropic polypeptide (GIP or gastric inhibitory polypeptide) is a 42-amino-acid hormone, secreted from the enteroendocrine K cells, which has insulin-releasing and extrapancreatic glucoregulatory actions. However, the unfavourable pharmacokinetic profile and the weak biological effects of native GIP limit its effectiveness for the treatment of type 2 diabetes. To overcome this, longer-acting GIP agonists exhibiting enzymatic stability and enhanced bioactivity have been generated and successfully tested in animal models of diabetes. Thus, GIP receptor agonists offer one of the newest classes of potential antidiabetic drug. GIP is also known to play a role in lipid metabolism and fat deposition. Accordingly, both genetic and chemical ablation of GIP signalling in mice with obesity-diabetes can protect against, or even reverse many of the obesity-associated metabolic disturbances. Strong parallels exist with the beneficial metabolic effects of Roux-en-Y gastric bypass in obese, insulin-resistant humans that surgically ablates GIP-secreting K cells. The purpose of this article is to highlight the therapeutic potential of GIP-based therapeutics in the treatment of type 2 diabetes and obesity.


Asunto(s)
Diabetes Mellitus Tipo 2/tratamiento farmacológico , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Obesidad/tratamiento farmacológico , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de la Hormona Gastrointestinal/antagonistas & inhibidores , Animales , Dipeptidil Peptidasa 4/metabolismo , Células Enteroendocrinas/metabolismo , Derivación Gástrica , Polipéptido Inhibidor Gástrico/fisiología , Polipéptido Inhibidor Gástrico/uso terapéutico , Humanos
18.
Endocrinology ; 150(5): 2118-26, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19179446

RESUMEN

The precise role of fat in postprandial glycemia and insulinemia has not been thoroughly researched because postprandial blood glucose and concurrent insulin secretion are largely assumed to be proportional to carbohydrate intake. Recent studies have suggested that dietary fat differentially regulates the postprandial insulin response. To explore this, we examined the effects of coadministered fat on glucose-induced glycemia and insulinemia in C57BL/6J mice. The insulin response to glucose was augmented by the addition of glycerol trioleate (TO) in a dose-dependent manner, which was associated with enhanced glucose transport from the circulation to muscle and adipose tissues. To investigate the mechanism underlying fat-induced hyperinsulinemia, we examined the release of the incretin hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1. TO increased GIP secretion, whereas glucagon-like peptide-1 secretion was unaffected. TO-induced hyperinsulinemia was significantly attenuated by the pretreatment of mice with a specific GIP antagonist. Diacylglycerol (DAG) promoted lower postprandial GIP and triglyceride responses and, when ingested with glucose, a lower insulin response compared with triacylglycerol of a similar fatty acid composition. Pluronic L-81, an inhibitor of chylomicron formation, reduced not only the triglyceride response but also TO-induced GIP secretion, indicating that the lower GIP response after DAG ingestion may be associated with retarded chylomicron formation in the small intestine. We conclude that dietary fat augments glucose-induced insulinemia via gut-derived GIP and, thereby, influences postprandial nutrient metabolism in mice. DAG promotes a lower GIP and thereby reduced insulin responses compared with triacylglycerol, which may differentially influence postprandial energy homeostasis.


Asunto(s)
Polipéptido Inhibidor Gástrico/fisiología , Glucosa/farmacología , Glicéridos/farmacología , Insulina/metabolismo , Administración Oral , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Grasas de la Dieta/farmacología , Combinación de Medicamentos , Ingestión de Alimentos/fisiología , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/metabolismo , Glucosa/administración & dosificación , Glucosa/farmacocinética , Glicéridos/administración & dosificación , Antagonistas de Hormonas/farmacología , Incretinas/metabolismo , Secreción de Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Fragmentos de Péptidos/farmacología , Receptores de la Hormona Gastrointestinal , Distribución Tisular
19.
Biol Chem ; 390(1): 75-80, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18937625

RESUMEN

Recent research suggests that long-term ablation of gastric inhibitory polypeptide (GIP) receptor signalling can reverse or prevent many of the metabolic abnormalities associated with dietary and genetically induced obesity-diabetes. The present study was designed to assess the sub-chronic effects of passive or active immunisation against GIP in ob/ob mice. Initial acute administration of GIP antibody together with oral glucose in ob/ob mice significantly increased the glycaemic excursion compared to controls (p<0.05). This was associated with a significant reduction (p<0.05) in the overall glucose-mediated insulin response. However, sub-chronic passive GIP immunisation was not associated with any changes in body weight, food intake or metabolic control. In contrast, active immunisation against GIP for 56 days in young ob/ob mice resulted in significantly (p<0.05) reduced circulating plasma glucose concentrations on day 56 compared to controls. There was a tendency for decreased circulating insulin in GIP immunised mice. The glycaemic response to intraperitoneal glucose was correspondingly improved (p<0.05) in mice immunised against GIP. Glucose-stimulated insulin levels were not significantly different from controls. Furthermore, insulin sensitivity was similar in mice immunised against GIP and respective controls. Overall, the results reveal that active, as opposed to passive, immunisation against GIP improves blood glucose control ob/ob mice.


Asunto(s)
Glucemia/metabolismo , Diabetes Mellitus/sangre , Modelos Animales de Enfermedad , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Polipéptido Inhibidor Gástrico/inmunología , Obesidad/sangre , Vacunación , Animales , Anticuerpos/inmunología , Diabetes Mellitus/metabolismo , Femenino , Polipéptido Inhibidor Gástrico/sangre , Homeostasis/inmunología , Humanos , Inmunización Pasiva , Insulina/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Factores de Tiempo
20.
Biochem Biophys Res Commun ; 376(1): 21-5, 2008 Nov 07.
Artículo en Inglés | MEDLINE | ID: mdl-18723001

RESUMEN

Gastric inhibitory polypeptide (GIP) is an incretin and directly promotes fat accumulation in adipocytes. Inhibition of GIP signaling prevents onset of obesity and increases fat oxidation in peripheral tissues under high-fat diet (HFD), but the mechanism is unknown. In the present study, we investigated the effects of inhibition of GIP signaling on adiponectin levels after 3 weeks of HFD by comparing wild-type (WT) mice and GIP receptor-deficient (Gipr(-/-)) mice. In HFD-fed Gipr(-/-) mice, fat oxidation was significantly increased and adiponectin mRNA levels in white adipose tissue and plasma adiponectin levels were significantly increased compared to those in HFD-fed WT mice. In addition, the PPARalpha mRNA level was increased and the ACC mRNA level was decreased in skeletal muscle of HFD-fed Gipr(-/-) mice compared with those in HFD-fed WT mice. These results indicate that inhibition of GIP signaling increases adiponectin levels, resulting in increased fat oxidation in peripheral tissues under HFD.


Asunto(s)
Adiponectina/metabolismo , Grasas de la Dieta/administración & dosificación , Polipéptido Inhibidor Gástrico/antagonistas & inhibidores , Adiponectina/sangre , Adiponectina/genética , Tejido Adiposo Blanco/metabolismo , Animales , Peso Corporal , Dieta , Polipéptido Inhibidor Gástrico/genética , Polipéptido Inhibidor Gástrico/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , PPAR alfa/genética , PPAR alfa/metabolismo , ARN Mensajero , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...