Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Endocrinol Metab ; 310(1): E81-90, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26530152

RESUMEN

Insulin resistance results in a compensatory increase in insulin secretion to maintain normoglycemia. Conversely, high insulin sensitivity results in reduced insulin secretion to prevent hypoglycemia. The mechanisms for this inverse adaptation are not well understood. We utilized highly insulin-sensitive mice, due to adipocyte-specific overexpression of the FOXC2 transcription factor, to study mechanisms of the reversed islet adaptation to increased insulin sensitivity. We found that Foxc2TG mice responded to mild hyperglycemia with insulin secretion significantly lower than that of wild-type mice; however, when severe hyperglycemia was induced, Foxc2TG mice demonstrated insulin secretion equal to or greater than that of wild-type mice. In response to autonomic nervous activation by 2-deoxyglucose, the acute suppression of insulin seen in wild-type mice was absent in Foxc2TG mice, suggesting impaired sympathetic signaling to the islet. Basal glucagon was increased in Foxc2TG mice, but they displayed severely impaired glucagon responses to cholinergic and autonomic nervous stimuli. These data suggest that the autonomic nerves contribute to the islet adaptation to high insulin sensitivity, which is compatible with a neuro-adipo regulation of islet function being instrumental for maintaining glucose regulation.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Sistema Nervioso Autónomo/fisiología , Glucagón/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Islotes Pancreáticos/citología , Animales , Forma de la Célula , Transdiferenciación Celular/fisiología , Femenino , Factores de Transcripción Forkhead/genética , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
2.
Diabetologia ; 57(9): 1876-83, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24939431

RESUMEN

AIMS/HYPOTHESIS: Inhibition of the enzyme dipeptidyl peptidase 4 (DPP-4), which cleaves and inactivates glucagon-like peptide 1 (GLP-1), is a glucose-lowering strategy in type 2 diabetes. Since DPP-4 is a ubiquitously distributed enzyme, we examined whether it is expressed in islets and whether an islet effect to inhibit DPP-4 may result in stimulated insulin secretion. METHODS: We investigated DPP-4 expression and activity in the islets of mouse models of obesity as well as human islets from non-diabetic and type 2 diabetic donors. We further investigated whether inhibition with DPP-4 inhibitors could promote insulin secretion via islet GLP-1 in isolated islets. RESULTS: DPP-4 was readily detected in mouse and human islets with species-specific cellular localisation. In mice, DPP-4 was expressed predominantly in beta cells, whereas in humans it was expressed nearly exclusively in alpha cells. DPP-4 activity was significantly increased in islets from diet-induced obese mice compared with mice fed a control diet. In humans, DPP-4 activity was significantly lower in islets from type 2 diabetic donors than in non-diabetic donors. In human islets, there was a significant positive correlation between DPP-4 activity and insulin secretory response to 16.7 mmol/l glucose. Treatment of mouse islets with the DPP-4 inhibitors, NVPDPP728 and vildagliptin, resulted in a significant potentiation of insulin secretion in a GLP-1-dependent manner, as this was inhibited by the GLP-1 receptor antagonist, Exendin (9-39), and was retained in glucose-dependent insulinotropic polypeptide (GIP) receptor-deficient mice but lost in mice lacking GLP-1 receptors or both incretin receptors. Human islets treated with the DPP-4 inhibitor, vildagliptin, showed increased secretion of insulin and intact GLP-1. CONCLUSIONS/INTERPRETATION: We conclude that DPP-4 is present and active in mouse and human islets, is regulated by the disease state, and that inhibition of islet DPP-4 activity can have direct effects on islet function. Inhibiting islet DPP-4 activity may therefore contribute to the insulin-secretory and glucose-lowering action of DPP-4 inhibition.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Dipeptidil Peptidasa 4/metabolismo , Islotes Pancreáticos/metabolismo , Adamantano/análogos & derivados , Adamantano/farmacología , Animales , Femenino , Glucosa/metabolismo , Humanos , Inmunohistoquímica , Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Nitrilos/farmacología , Pirrolidinas/farmacología , Vildagliptina
3.
Biochem Biophys Res Commun ; 453(3): 398-404, 2014 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-25268763

RESUMEN

Dipeptidyl peptidase 4 (DPP-4) was recently found to be expressed in human and mouse islets with different expression patterns. However, whether species-dependent expression pattern is a generalized phenomenon and whether islet DPP-4 activity is regulated are not known. This study was conducted to investigate DPP-4 localization in several different species, and to examine the impact of glucose, incretin hormones, and insulin on islet DPP-4 activity. It was shown by immuofluorescent staining that there were two distinct species-specific expression patterns of islet DPP-4. The enzyme was expressed exclusively in α-cells in human and pig islets, but primarily in ß-cells in mouse and rat islets. INS-1 832/13 cells also expressed DPP-4, and inhibition of DPP-4 enhanced insulin secretion in the presence of glucagon-like peptide-1 (GLP-1) in the cells. DPP-4 activity was remarkably robust when cultured with high glucose, incretin hormones, and insulin in mouse and human islets as well as INS-1 832/13 cells and islet DPP-4 activity and expression pattern was not altered in double incretin receptor knockout mice, compared to wild type mice. We conclude that islet DPP-4 is species-specifically expressed in α-cell and ß-cell dominant patterns in several species and both patterns remained robust in enzyme activity during short-term metabolic challenge.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Islotes Pancreáticos/enzimología , Animales , Femenino , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratas
4.
Biochem Biophys Res Commun ; 425(4): 812-7, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22892131

RESUMEN

The incretin - glucose-dependent insulinotropic polypeptide (GIP) - and the pro-inflammatory cytokine osteopontin are known to have important roles in the regulation of adipose tissue functions. In this work we show that GIP stimulates lipogenesis and osteopontin expression in primary adipocytes. The GIP-induced increase in osteopontin expression was inhibited by the NFAT (the transcription factor nuclear factor of activated T-cells) inhibitor A-285222. Also, the NFAT kinase glycogen synthase kinase (GSK) 3 was upregulated by GIP. To test whether cAMP might be involved in GIP-mediated effects on osteopontin a number of strategies were used. Thus, the ß3-adrenergic receptor agonist CL316,243 stimulated osteopontin expression, an effects which was mimicked by OPC3911, a specific inhibitor of phosphodiesterase 3. Furthermore, treatment of phosphodiesterase 3B knock-out mice with CL316,243 resulted in a dramatic upregulation of osteopontin in adipose tissue which was not the case in wild-type mice. In summary, we delineate mechanisms by which GIP stimulates osteopontin in adipocytes. Given the established link between osteopontin and insulin resistance, our data suggest that GIP by stimulating osteopontin expression, also could promote insulin resistance in adipocytes.


Asunto(s)
Adipocitos/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Polipéptido Inhibidor Gástrico/fisiología , Lipogénesis/fisiología , Factores de Transcripción NFATC/fisiología , Osteopontina/metabolismo , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Polipéptido Inhibidor Gástrico/farmacología , Insulina/metabolismo , Lipogénesis/efectos de los fármacos , Masculino , Ratones , Factores de Transcripción NFATC/antagonistas & inhibidores , Osteopontina/genética , Pirazoles/farmacología , Ratas , Ratas Sprague-Dawley
5.
Pan Afr Med J ; 37: 97, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33425130

RESUMEN

A 50-year-old woman presented with a 5-month history of recurrent urinary tract infections. She had no complaints of any intestinal symptoms. She had been treated previously with oral antibiotics. The episodes became more frequent and she started with pain in the lower abdomen and fetid urine. Complete study lead to diagnosis of adenocarcinoma of the appendix with bladder fistula. The lesion was removed by laparoscopic right hemicolectomy and en bloc partial cystectomy. Pathological examination revealed a mucinous adenocarcinoma that had originated in the appendix and extended into the bladder wall. Six years after the operation, the patient remains asymptomatic with no evidence of recurrent or metastatic disease. Appendiceal carcinoma extending to the bladder is extremely rare and approximately 40 cases have been described. Management of recurrent urinary tract infections should not limit to empiric antibiotic therapy before the exclusion of possible organic causes. Appendiceal carcinoma may invade the bladder without intestinal symptoms but with urinary symptoms only, because of its anatomical position. The recommended treatment for non-carcinoid appendiceal tumours is right hemicolectomy and for T4 tumours en bloc resection of the involved structures. Further study is needed to determine adjuvant therapy. A literature review was made.


Asunto(s)
Adenocarcinoma Mucinoso/diagnóstico , Neoplasias del Apéndice/diagnóstico , Fístula de la Vejiga Urinaria/diagnóstico , Infecciones Urinarias/etiología , Adenocarcinoma Mucinoso/complicaciones , Adenocarcinoma Mucinoso/cirugía , Neoplasias del Apéndice/complicaciones , Neoplasias del Apéndice/cirugía , Colectomía , Cistectomía , Femenino , Humanos , Laparoscopía , Persona de Mediana Edad , Fístula de la Vejiga Urinaria/complicaciones , Fístula de la Vejiga Urinaria/cirugía
6.
Sci Rep ; 10(1): 17257, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33057063

RESUMEN

Neuregulin protein 1 (NRG1) is a large (> 60-amino-acid) natural peptide ligand for the ErbB protein family members HER3 and HER4. We developed an agonistic antibody modality, termed antibody ligand mimetics (ALM), by incorporating complex ligand agonists such as NRG1 into an antibody scaffold. We optimized the linker and ligand length to achieve native ligand activity in HEK293 cells and cardiomyocytes derived from induced pluripotent stem cells (iPSCs) and used a monomeric Fc-ligand fusion platform to steer the ligand specificity toward HER4-dominant agonism. With the help of selectivity engineering, these enhanced ALM molecules can provide an antibody scaffold with increased receptor specificity and the potential to greatly improve the pharmacokinetics, stability, and downstream developability profiles from the natural ligand approach. This ligand mimetic design and optimization approach can be expanded to apply to other cardiovascular disease targets and emerging therapeutic areas, providing differentiated drug molecules with increased specificity and extended half-life.


Asunto(s)
Anticuerpos Monoclonales/química , Neurregulina-1/química , Receptor ErbB-4/agonistas , Anticuerpos Monoclonales/metabolismo , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/química , Células Madre Pluripotentes Inducidas/metabolismo , Cinética , Ligandos , Miocitos Cardíacos/química , Miocitos Cardíacos/metabolismo , Neurregulina-1/metabolismo , Unión Proteica , Receptor ErbB-4/metabolismo , Transducción de Señal
7.
Mol Cell Endocrinol ; 473: 186-193, 2018 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-29409957

RESUMEN

It has been reported that the incretin system, including regulated GLP-1 secretion and locally expressed DPP-4, is present in pancreatic islets. In this study we comprehensively evaluated the expression and role of DPP-4 in islet alpha and beta cells from non-diabetic (ND) and type 2 diabetic (T2D) individuals, including the effects of its inhibition on beta cell function and survival. Isolated islets were prepared from 25 ND and 18 T2D organ donors; studies were also performed with the human insulin-producing EndoC-ßH1 cells. Morphological (including confocal microscopy), ultrastructural (electron microscopy, EM), functional (glucose-stimulated insulin secretion), survival (EM and nuclear dyes) and molecular (RNAseq, qPCR and western blot) studies were performed under several different experimental conditions. DPP-4 co-localized with glucagon and was also expressed in human islet insulin-containing cells. Furthermore, DPP-4 was expressed in EndoC-ßH1 cells. The proportions of DPP-4 positive alpha and beta cells and DPP-4 gene expression were significantly lower in T2D islets. A DPP-4 inhibitor protected ND human beta cells and EndoC-ßH1 cells against cytokine-induced toxicity, which was at least in part independent from GLP1 and associated with reduced NFKB1 expression. Finally, DPP-4 inhibition augmented glucose-stimulated insulin secretion, reduced apoptosis and improved ultrastructure in T2D beta cells. These results demonstrate the presence of DPP-4 in human islet alpha and beta cells, with reduced expression in T2D islets, and show that DPP-4 inhibition has beneficial effects on human ND and T2D beta cells. This suggests that DPP-4, besides playing a role in incretin effects, directly affects beta cell function and survival.


Asunto(s)
Diabetes Mellitus Tipo 2/enzimología , Diabetes Mellitus Tipo 2/patología , Dipeptidil Peptidasa 4/metabolismo , Células Secretoras de Insulina/enzimología , Anciano , Apoptosis/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Citocinas/toxicidad , Citoprotección/efectos de los fármacos , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Humanos , Secreción de Insulina/efectos de los fármacos , Células Secretoras de Insulina/ultraestructura , Masculino , Persona de Mediana Edad
8.
Eur J Pharmacol ; 780: 46-52, 2016 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26997369

RESUMEN

It has been suggested that neural mechanisms may contribute to effects of the incretin hormones, and, therefore, also to the effects of dipeptidyl peptidase (DPP-4) inhibition. We therefore examined whether muscarinic mechanisms are involved in the stimulation of insulin secretion by DPP-4 inhibition. Fasted, anesthetized mice were given intraperitoneal saline or the muscarinic antagonist atropine (5mg/kg) before duodenal glucose (75mg/mouse), with or without the DPP-4 inhibitor NVPDPP728 (0.095mg/mouse), or before intravenous glucose (0.35g/kg) with or without co-administration with GLP-1 or glucose-dependent insulinotropic polypeptide (GIP) (both 3nmol/kg). Furthermore, isolated islets were incubated (1h) in 2.8 and 11.1mM glucose, with or without GIP or GLP-1 (both 100nM), in the presence or absence of atropine (100µM). Duodenal glucose increased circulating insulin and this effect was potentiated by DPP-4 inhibition. The increase in insulin achieved by DPP-4 inhibition was reduced by atropine by approximately 35%. Duodenal glucose also elicited an increase in circulating intact GLP-1 and GIP and this was augmented by DPP-4 inhibition, but these effects were not affected by atropine. Atropine did also not affect the augmentation by GLP-1 and GIP on glucose-stimulated insulin secretion from isolated islets. Based on these findings, we suggest that muscarinic mechanisms contribute to the stimulation of insulin secretion by DPP-4 inhibition through neural effects induced by GLP-1 and GIP whereas neural effects do not affect the levels of GLP-1 or GIP or the islet effects of the two incretin hormones.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Islotes Pancreáticos/efectos de los fármacos , Fenómenos Fisiológicos del Sistema Nervioso , Animales , Carbacol/farmacología , Femenino , Glucosa/administración & dosificación , Glucosa/farmacología , Incretinas/metabolismo , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Antagonistas Muscarínicos/farmacología , Fenómenos Fisiológicos del Sistema Nervioso/efectos de los fármacos , Receptores Muscarínicos/metabolismo
9.
Peptides ; 79: 58-65, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27020250

RESUMEN

The incretin hormones, glucose dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1), potentiate insulin secretion and are responsible for the majority of insulin secretion that occurs after a meal. They may also, however, have a fundamental role in pancreatic beta cell development and function, independently of their role in potentiating insulin secretion after a meal. This has led to observations that a loss of GIP or GLP-1 action affects normal beta cell function, however each one of the incretin hormones may compensate when the action of the other is lost and therefore the overall impact of the incretin hormones on beta cell function is not known. We therefore utilized a mouse line deficient in both the GLP-1 and GIP receptor genes, the double incretin receptor knockout (DIRKO), to determine the consequences of a lifelong, complete lack of incretin hormone action on beta cell function, in vivo, in intact animals. We found that DIRKO mice displayed impaired glucose tolerance and insulin secretion in response to both oral glucose and mixed meal tolerance tests compared to wild-type mice. Assessment of beta cell function using the hyperglycemic clamp technique revealed an 80% decrease in first phase insulin response in DIRKO mice, but a normal second phase insulin secretion. A similar decline was seen when wild-type mice were given acute intravenous injection of glucose together with the GLP-1 receptor antagonist Ex9-39. Ex vivo assessments of the pancreas revealed significantly fewer islets in the pancreata of DIRKO mice despite no differences in total pancreatic mass. Insulin secretion from isolated islets of DIRKO mice was impaired to a similar extent to that seen during the hyperglycemic clamp. Insulin secretion in wild-type islets was impaired by acute treatment with Ex9-39 to a similar extent as the in vivo intravenous glucose tolerance tests. In conclusion, a loss of the action of both incretin hormones results in direct impairment of beta cell function both in vivo and in vitro in a process that appears to be independent of the intestinally secreted incretin hormones. We therefore conclude that the incretin hormones together significantly impact both beta-cell function and beta-cell development.


Asunto(s)
Receptor del Péptido 1 Similar al Glucagón/genética , Células Secretoras de Insulina/fisiología , Receptores de la Hormona Gastrointestinal/genética , Animales , Glucemia , Peso Corporal , Células Cultivadas , Femenino , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Insulina/sangre , Insulina/metabolismo , Secreción de Insulina , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de los Órganos , Páncreas/citología , Páncreas/crecimiento & desarrollo , Receptores de la Hormona Gastrointestinal/metabolismo
10.
J Endocrinol ; 228(3): 171-8, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26698567

RESUMEN

Stimulation of insulin secretion by short-term glucagon receptor (GCGR) activation is well characterized; however, the effect of long-term GCGR activation on ß-cell function is not known, but of interest, since hyperglucagonemia occurs early during development of type 2 diabetes. Therefore, we examined whether chronic GCGR activation affects insulin secretion in glucose intolerant mice. To induce chronic GCGR activation, high-fat diet fed mice were continuously (2 weeks) infused with the stable glucagon analog ZP-GA-1 and challenged with oral glucose and intravenous glucose±glucagon-like peptide 1 (GLP1). Islets were isolated to evaluate the insulin secretory response to glucose±GLP1 and their pancreas were collected for immunohistochemical analysis. Two weeks of ZP-GA-1 infusion reduced insulin secretion both after oral and intravenous glucose challenges in vivo and in isolated islets. These inhibitory effects were corrected for by GLP1. Also, we observed increased ß-cell area and islet size. We conclude that induction of chronic ZP-GA-1 levels in glucose intolerant mice markedly reduces insulin secretion, and thus, we suggest that chronic activation of the GCGR may contribute to the failure of ß-cell function during development of type 2 diabetes.


Asunto(s)
Intolerancia a la Glucosa/fisiopatología , Insulina/metabolismo , Receptores de Glucagón/fisiología , Animales , Diabetes Mellitus Tipo 2/fisiopatología , Dieta Alta en Grasa , Femenino , Glucagón/administración & dosificación , Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/administración & dosificación , Glucosa/administración & dosificación , Intolerancia a la Glucosa/etiología , Prueba de Tolerancia a la Glucosa/métodos , Secreción de Insulina , Células Secretoras de Insulina/fisiología , Ratones , Ratones Endogámicos C57BL , Receptores de Glucagón/efectos de los fármacos
11.
Eur J Pharmacol ; 764: 189-194, 2015 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-26144370

RESUMEN

In type 1 diabetes, there is a rapid loss of glycemic control immediately after onset of the disease. We aimed to determine if the deterioration of glycemic control that occurs early after the onset of insulin-deficient diabetes could be blunted by treatment with recombinant fibroblast growth factor 21 (FGF21). Normal C57BL/6J mice made diabetic by a single high dose injection of streptozotocin (STZ) were randomized to receive twice daily subcutaneous injection of vehicle or recombinant human FGF21 at doses of 0.3 and 1.0 mg/kg for 10 days. Body weight was recorded daily and 5 h fasted glucose, insulin, glucagon, free fatty acids and ketones were determined at 6 and 10 days post-randomization. The increase in fasting plasma glucose induced by STZ in untreated mice was prevented with FGF21 at 0.3 mg/kg BID. In contrast, at 1.0 mg/kg BID, FGF21 did not prevent the rise in plasma glucose after STZ. At the end of the study, plasma glucagon was significantly higher in the diabetic group treated with FGF21 1.0 mg/kg BID than in the untreated group. This was not seen for the group treated with FGF21 0.3 mg/kg BID. There were significant dose dependent reductions in plasma free fatty acids with FGF21 treatment but no significant change in plasma ketones (ß-hydroxybutyrate). FGF21 treatment did not have significant effects on body weight in lean insulin deficient mice. In conclusion, FGF21 prevents increases in glycaemia and has lipid lowering properties in mouse models of insulin deficient diabetes, although by increasing the dose increased glucagon levels are seen and hyperglycemia persists.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Factores de Crecimiento de Fibroblastos/uso terapéutico , Tejido Adiposo/metabolismo , Animales , Glucemia/análisis , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/metabolismo , Femenino , Factores de Crecimiento de Fibroblastos/farmacología , Glucagón/sangre , Insulina/sangre , Ratones Endogámicos C57BL , Obesidad/sangre , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Páncreas/metabolismo
12.
J Clin Endocrinol Metab ; 100(2): 561-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25375983

RESUMEN

CONTEXT: Postprandial glucose homeostasis is regulated through the secretion of glucagon-like peptide 1 (GLP-1) through the stimulation of insulin secretion and inhibition of glucagon secretion. However, how these processes dynamically adapt to demands created by caloric challenges achieved during daily life is not known. OBJECTIVE: The objective of the study was to explore the adaptation of incretin and islet hormones after mixed meals of increasing size in healthy subjects. DESIGN: Twenty-four healthy lean subjects ingested a standard breakfast after an overnight fast followed, after 4 hours, by a lunch of a different size (511, 743, and 1034 kcal) but with identical nutrient composition together with 1.5 g paracetamol. Glucose, insulin, C-peptide, glucagon, intact GLP-1, and glucose-dependent insulinotropic polypeptide (GIP) and paracetamol were measured after the meals. MAIN OUTCOME MEASURE: Area under the 180-minute curve (AUC) for insulin, C-peptide, glucagon, GLP-1, and GIP and model-derived ß-cell function and paracetamol appearance were calculated. RESULTS: Glucose profiles were similar after the two larger meals, whereas after the smaller meal, there was a postpeak reduction below baseline to a nadir of 3.8 ± 0.1 mmol/L after 75 minutes (P < .001). The AUC for GLP-1, GIP, insulin, and C-peptide were significantly higher by increasing the caloric load as was ß-cell sensitivity to glucose. In contrast, the AUC glucagon was the same for all three meals, although there was an increase in glucagon after the postpeak glucose reduction in the smaller meal. The 0- to 20-minute paracetamol appearance was increased by increasing meal size. CONCLUSION: Mixed lunch meals of increasing size elicit a caloric-dependent insulin response due to increased ß-cell secretion achieved by increased GIP and GLP-1 levels. The adaptation at larger meals results in identical glucose excursions, whereas after a lower caloric lunch, the insulin response is high, resulting in a postpeak suppression of glucose below baseline.


Asunto(s)
Polipéptido Inhibidor Gástrico/sangre , Péptido 1 Similar al Glucagón/sangre , Comidas , Periodo Posprandial/fisiología , Adulto , Glucemia/metabolismo , Péptido C/metabolismo , Femenino , Glucagón/sangre , Voluntarios Sanos , Humanos , Insulina/sangre , Masculino , Adulto Joven
13.
Biochem Pharmacol ; 96(3): 267-77, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26049045

RESUMEN

We report the production and metabolic phenotype of a mouse line in which the Fmo5 gene is disrupted. In comparison with wild-type (WT) mice, Fmo5(-/-) mice exhibit a lean phenotype, which is age-related, becoming apparent after 20 weeks of age. Despite greater food intake, Fmo5(-/-) mice weigh less, store less fat in white adipose tissue (WAT), have lower plasma glucose and cholesterol concentrations and enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure, with no increase in physical activity. An increase in respiratory exchange ratio during the dark phase, the period in which the mice are active, indicates a switch from fat to carbohydrate oxidation. In comparison with WT mice, the rate of fatty acid oxidation in Fmo5(-/-) mice is higher in WAT, which would contribute to depletion of lipid stores in this tissue, and lower in skeletal muscle. Five proteins were down regulated in the liver of Fmo5(-/-) mice: aldolase B, ketohexokinase and cytosolic glycerol 3-phosphate dehydrogenase (GPD1) are involved in glucose or fructose metabolism and GPD1 also in production of glycerol 3-phosphate, a precursor of triglyceride biosynthesis; HMG-CoA synthase 1 is involved in cholesterol biosynthesis; and malic enzyme 1 catalyzes the oxidative decarboxylation of malate to pyruvate, in the process producing NADPH for use in lipid and cholesterol biosynthesis. Down regulation of these proteins provides a potential explanation for the reduced fat deposits and lower plasma cholesterol characteristic of Fmo5(-/-) mice. Our results indicate that disruption of the Fmo5 gene slows metabolic ageing via pleiotropic effects.


Asunto(s)
Tejido Adiposo Blanco/enzimología , Envejecimiento/genética , Efecto Fundador , Regulación de la Expresión Génica , Oxigenasas/genética , Envejecimiento/metabolismo , Animales , Glucemia/metabolismo , Peso Corporal/genética , Colesterol/sangre , Metabolismo Energético/genética , Fructoquinasas/genética , Fructoquinasas/metabolismo , Fructosa-Bifosfato Aldolasa/genética , Fructosa-Bifosfato Aldolasa/metabolismo , Genotipo , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/genética , Glicerol-3-Fosfato Deshidrogenasa (NAD+)/metabolismo , Hidroximetilglutaril-CoA Sintasa/genética , Hidroximetilglutaril-CoA Sintasa/metabolismo , Metabolismo de los Lípidos/genética , Hígado/enzimología , Malato Deshidrogenasa/genética , Malato Deshidrogenasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/enzimología , Oxidación-Reducción , Oxigenasas/deficiencia , Fenotipo
14.
Diabetes ; 63(7): 2196-202, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24962916

RESUMEN

Dipeptidyl peptidase (DPP)-4 inhibition is a glucose-lowering treatment for type 2 diabetes. The classical mechanism for DPP-4 inhibitors is that they inhibit DPP-4 activity in peripheral plasma, which prevents the inactivation of the incretin hormone glucagon-like peptide (GLP)-1 in the peripheral circulation. This in turn increases circulating intact GLP-1, which results in stimulated insulin secretion and inhibited glucagon secretion, in turn increasing glucose utilization and diminishing hepatic glucose production, which, through reduction in postprandial and fasting glucose, reduces HbA1c. However, recent experimental studies in mainly rodents but also to a limited degree in humans have found additional mechanisms for DPP-4 inhibitors that may contribute to their glucose-lowering action. These nonclassical mechanisms include 1) inhibition of gut DPP-4 activity, which prevents inactivation of newly released GLP-1, which in turn augments GLP-1-induced activations of autonomic nerves and results in high portal GLP-1 levels, resulting in inhibited glucose production through portal GLP-1 receptors; 2) inhibition of islet DPP-4 activity, which prevents inactivation of locally produced intact GLP-1 in the islets, thereby augmenting insulin secretion and inhibiting glucagon secretion and possibly preventing islet inflammation; and 3) prevention of the inactivation of other bioactive peptides apart from GLP-1, such as glucose-dependent insulinotropic polypeptide, stromal-derived factor-1α, and pituitary adenylate cyclase-activating polypeptide, which may improve islet function. These pleiotropic effects may contribute to the effects of DPP-4 inhibition. This Perspectives in Diabetes outlines and discusses these nonclassical mechanisms of DPP-4 inhibition.


Asunto(s)
Glucemia/efectos de los fármacos , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Hipoglucemiantes/farmacología , Diabetes Mellitus Tipo 2/sangre , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/farmacología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Transducción de Señal/efectos de los fármacos
15.
Metabolism ; 63(12): 1568-76, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25301258

RESUMEN

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.


Asunto(s)
Células Secretoras de Insulina/metabolismo , Receptores de Glucagón/biosíntesis , Animales , Arginina/farmacología , Peso Corporal/efectos de los fármacos , Carbacol/farmacología , Glucagón/farmacología , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/farmacología , Prueba de Tolerancia a la Glucosa , Humanos , Hipoglucemiantes/farmacología , Insulina/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones , Ratones Transgénicos , Receptores de Glucagón/efectos de los fármacos , Receptores de Glucagón/genética , Receptores Muscarínicos/efectos de los fármacos
16.
Physiol Rep ; 2(5)2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24843074

RESUMEN

Insulin sensitivity declines in overweight and obese individuals and, under normal conditions, insulin secretion adaptively increases which in healthy non-diabetic subjects maintains normal glycemia. This adaptation is best described by the disposition index derived from modeling of insulin and glucose data from an intravenous glucose tolerance testing (IVGTT). One caveat of the IVGTT is that basing the glucose dose on the individual total body weight can result in large differences in the amount of glucose given to lean and obese individuals. The effect this has on determination of insulin sensitivity and ß-cell function is unknown. In this study, we therefore evaluated alternative glucose dosing regimens for determination of the impact of glucose dosing on measures of ß-cell function in normal and diet-induced obese (DIO) mice. The glucose dosing regimens used for the IVGTT were 0.35 mg per kg total body weight (BW) or per kg lean BW or a fixed glucose dose based on the average BW for all experimental mice. Each regimen detected a similar decrease in insulin sensitivity in DIO mice. The different glucose dosing regimens gave, however, diverging results in regard to glucose elimination and the acute insulin response. Thus, the fixed-dose regimen was the only that revealed impairment of glucose elimination, whereas dosing according to total BW was the only regimen which showed significant increases in acute insulin response in DIO mice. The fixed-dose glucose dosing regimen was the only that revealed a significant decline in the disposition index value in DIO mice, which is characteristic of type 2 diabetes in humans. Our results therefore show that using different glucose dosing regimens during IVGTT in DIO mice one can model different aspects of physiology which are similar to prediabetes and type 2 diabetes in humans, with the fixed-dose regimen producing a phenotype that most closely resembles human type 2 diabetes.

17.
Diabetes ; 63(1): 101-10, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24062250

RESUMEN

Mice genetically deficient in the glucagon receptor (Gcgr(-/-)) show improved glucose tolerance, insulin sensitivity, and α-cell hyperplasia. In addition, Gcgr(-/-) mice do not develop diabetes after chemical destruction of ß-cells. Since fibroblast growth factor 21 (FGF21) has insulin-independent glucose-lowering properties, we investigated whether FGF21 was contributing to diabetes resistance in insulin-deficient Gcgr(-/-) mice. Plasma FGF21 was 25-fold higher in Gcgr(-/-) mice than in wild-type mice. FGF21 was found to be expressed in pancreatic ß- and α-cells, with high expression in the hyperplastic α-cells of Gcgr(-/-) mice. FGF21 expression was also significantly increased in liver and adipose tissue of Gcgr(-/-) mice. To investigate the potential antidiabetic actions of FGF21 in insulin-deficient Gcgr(-/-) mice, an FGF21-neutralizing antibody was administered prior to oral glucose tolerance tests (OGTTs). FGF21 neutralization caused a decline in glucose tolerance in insulin-deficient Gcgr(-/-) mice during the OGTT. Despite this decline, insulin-deficient Gcgr(-/-) mice did not develop hyperglycemia. Glucagon-like peptide 1 (GLP-1) also has insulin-independent glucose-lowering properties, and an elevated circulating level of GLP-1 is a known characteristic of Gcgr(-/-) mice. Neutralization of FGF21, while concurrently blocking the GLP-1 receptor with the antagonist Exendin 9-39 (Ex9-39), resulted in significant hyperglycemia in insulin-deficient Gcgr(-/-) mice, while blocking with Ex9-39 alone did not. In conclusion, FGF21 acts additively with GLP-1 to prevent insulinopenic diabetes in mice lacking glucagon action.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Hiperglucemia/metabolismo , Receptores de Glucagón/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Receptor del Péptido 1 Similar al Glucagón , Células Secretoras de Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Hiperglucemia/genética , Células Secretoras de Insulina/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados , Páncreas/metabolismo , Fragmentos de Péptidos/farmacología , Receptores de Glucagón/antagonistas & inhibidores , Receptores de Glucagón/genética
18.
Biochem Pharmacol ; 90(1): 88-95, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24792439

RESUMEN

Flavin-containing monooxygenases (FMOs) of mammals are thought to be involved exclusively in the metabolism of foreign chemicals. Here, we report the unexpected finding that mice lacking Fmos 1, 2 and 4 exhibit a lean phenotype and, despite similar food intake, weigh less and store less triglyceride in white adipose tissue (WAT) than wild-type mice. This is a consequence of enhanced whole-body energy expenditure, due mostly to increased resting energy expenditure (REE). This is fuelled, in part, by increased fatty acid ß-oxidation in skeletal muscle, which would contribute to depletion of lipid stores in WAT. The enhanced energy expenditure is attributed, in part, to an increased capacity for exercise. There is no evidence that the enhanced REE is due to increased adaptive thermogenesis; instead, our results are consistent with the operation in WAT of a futile energy cycle. In contrast to FMO2 and FMO4, FMO1 is highly expressed in metabolic tissues, including liver, kidney, WAT and BAT. This and other evidence implicates FMO1 as underlying the phenotype. The identification of a novel, previously unsuspected, role for FMO1 as a regulator of energy homeostasis establishes, for the first time, a role for a mammalian FMO in endogenous metabolism. Thus, FMO1 can no longer be considered to function exclusively as a xenobiotic-metabolizing enzyme. Consequently, chronic administration of drugs that are substrates for FMO1 would be expected to affect energy homeostasis, via competition for endogenous substrates, and, thus, have important implications for the general health of patients and their response to drug therapy.


Asunto(s)
Metabolismo Energético/genética , Regulación de la Expresión Génica , Oxigenasas/genética , Oxigenasas/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adiposidad/genética , Animales , Peso Corporal/genética , Ácidos Grasos/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Actividad Motora , Músculo Esquelético/metabolismo , Oxidación-Reducción , Consumo de Oxígeno/genética , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
19.
Diabetes ; 62(6): 2088-94, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23349498

RESUMEN

Low-grade inflammation in obesity is associated with accumulation of the macrophage-derived cytokine osteopontin (OPN) in adipose tissue and induction of local as well as systemic insulin resistance. Since glucose-dependent insulinotropic polypeptide (GIP) is a strong stimulator of adipogenesis and may play a role in the development of obesity, we explored whether GIP directly would stimulate OPN expression in adipose tissue and thereby induce insulin resistance. GIP stimulated OPN protein expression in a dose-dependent fashion in rat primary adipocytes. The level of OPN mRNA was higher in adipose tissue of obese individuals (0.13 ± 0.04 vs. 0.04 ± 0.01, P < 0.05) and correlated inversely with measures of insulin sensitivity (r = -0.24, P = 0.001). A common variant of the GIP receptor (GIPR) (rs10423928) gene was associated with a lower amount of the exon 9-containing isoform required for transmembrane activity. Carriers of the A allele with a reduced receptor function showed lower adipose tissue OPN mRNA levels and better insulin sensitivity. Together, these data suggest a role for GIP not only as an incretin hormone but also as a trigger of inflammation and insulin resistance in adipose tissue. Carriers of the GIPR rs10423928 A allele showed protective properties via reduced GIP effects. Identification of this unprecedented link between GIP and OPN in adipose tissue might open new avenues for therapeutic interventions.


Asunto(s)
Tejido Adiposo/metabolismo , Polipéptido Inhibidor Gástrico/farmacología , Resistencia a la Insulina/fisiología , Tejido Adiposo/efectos de los fármacos , Adolescente , Adulto , Anciano , Alelos , Animales , Células Cultivadas , Femenino , Humanos , Técnicas In Vitro , Resistencia a la Insulina/genética , Masculino , Ratones , Persona de Mediana Edad , Osteopontina/genética , Ratas , Receptores de la Hormona Gastrointestinal/genética , Receptores de la Hormona Gastrointestinal/metabolismo , Adulto Joven
20.
Nutrients ; 4(10): 1367-81, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-23201760

RESUMEN

BACKGROUND: The C57BL/6 mouse fed a high fat diet is a common and valuable model in experimental studies of obesity and type 2 diabetes (T2D). Different high fat diets are used and in order to determine which diet produces a model most accurately resembling human T2D, they need to be compared head-to-head. METHODS: Four different diets, the 60% high fat diet (HFD) and the 58% high fat-high sucrose Surwit diet (HFHS) and their respective controls, were compared in C57BL/6J mice using glucose tolerance tests (IVGTT) and the euglycemic clamp. RESULTS: Mice fed a HFD gained more weight than HFHS fed mice despite having similar energy intake. Both high fat diet models were glucose intolerant after eight weeks. Mice fed the HFD had elevated basal insulin, which was not seen in the HFHS group. The acute insulin response (AIR) was unchanged in the HFD group, but slightly increased in the HFHS diet group. The HFHS diet group had a threefold greater total insulin secretion during the IVGTT compared to its control, while no differences were seen in the HFD group. Insulin sensitivity was decreased fourfold in the HFD group, but not in the HFHS diet group. CONCLUSION: The HFD and HFHS diet models show differential effects on the development of insulin resistance and beta cell adaptation. These discrepancies are important to acknowledge in order to select the appropriate diet for specific studies.


Asunto(s)
Diabetes Mellitus Tipo 2/etiología , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/efectos adversos , Sacarosa en la Dieta/efectos adversos , Modelos Animales de Enfermedad , Islotes Pancreáticos/efectos de los fármacos , Obesidad/etiología , Animales , Diabetes Mellitus Tipo 2/metabolismo , Ingestión de Energía , Femenino , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/terapia , Insulina/metabolismo , Resistencia a la Insulina , Secreción de Insulina , Islotes Pancreáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Aumento de Peso/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA