Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Diabetologia ; 58(9): 2124-32, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26049402

RESUMEN

AIMS/HYPOTHESIS: We assessed the contribution of glucagon-like peptide-1 (GLP-1) receptor (GLP-1R) signalling to thermogenesis induced by high-fat diet (HFD) consumption. Furthermore, we determined whether brown adipose tissue (BAT) activity contributes to weight loss induced by chronic subcutaneous treatment with the GLP-1R agonist, liraglutide, in a model of diet-induced obesity. METHODS: Metabolic phenotyping was performed using indirect calorimetry in wild-type (WT) and Glp1r-knockout (KO) mice during chow and HFD feeding at room temperature and at thermoneutrality. In a separate study, we investigated the contribution of BAT thermogenic capacity to the weight lowering effect induced by GLP-1 mimetics by administering liraglutide (10 or 30 nmol kg(-1) day(-1) s.c.) to diet-induced obese (DIO) mice for 6 or 4 weeks, respectively. In both studies, animals were subjected to a noradrenaline (norepinephrine)-stimulated oxygen consumption [Formula: see text] test. RESULTS: At thermoneutrality, HFD-fed Glp1r-KO mice had similar energy expenditure (EE) compared with HFD-fed WT controls. However, HFD-fed Glp1r-KO mice exhibited relatively less EE when housed at a cooler standard room temperature, and had relatively lower [Formula: see text] in response to a noradrenaline challenge, which is consistent with impaired BAT thermogenic capacity. In contrast to the loss of function model, chronic peripheral liraglutide treatment did not increase BAT activity as determined by noradrenaline-stimulated [Formula: see text] and BAT gene expression. CONCLUSIONS/INTERPRETATION: These data suggest that although endogenous GLP-1R signalling contributes to increased BAT thermogenesis, this mechanism does not play a significant role in the food intake-independent body weight lowering effect of the GLP-1 mimetic liraglutide in DIO mice.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Animales , Composición Corporal , Calorimetría Indirecta , Dieta , Dieta Alta en Grasa , Ingestión de Alimentos , Metabolismo Energético/fisiología , Liraglutida/química , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Obesos , Norepinefrina/química , Consumo de Oxígeno , Fenotipo , Transducción de Señal , Temperatura , Termogénesis
2.
Gut ; 63(8): 1238-46, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24107591

RESUMEN

OBJECTIVE: Surgical interventions that prevent nutrient exposure to the duodenum are among the most successful treatments for obesity and diabetes. However, these interventions are highly invasive, irreversible and often carry significant risk. The duodenal-endoluminal sleeve (DES) is a flexible tube that acts as a barrier to nutrient-tissue interaction along the duodenum. We implanted this device in Zucker Diabetic Fatty (ZDF) rats to gain greater understanding of duodenal nutrient exclusion on glucose homeostasis. DESIGN: ZDF rats were randomised to four groups: Naive, sham ad libitum, sham pair-fed, and DES implanted. Food intake, body weight (BW) and body composition were measured for 28 days postoperatively. Glucose, lipid and bile acid metabolism were evaluated, as well as histological assessment of the upper intestine. RESULTS: DES implantation induced a sustained decrease in BW throughout the study that was matched by pair-fed sham animals. Decreased BW resulted from loss of fat, but not lean mass. DES rats were also found to be more glucose tolerant than either ad libitum-fed or pair-fed sham controls, suggesting fat mass independent metabolic benefits. DES also reduced circulating triglyceride and glycerol levels while increasing circulating bile acids. Interestingly, DES stimulated a considerable increase in villus length throughout the upper intestine, which may contribute to metabolic improvements. CONCLUSIONS: Our preclinical results validate DES as a promising therapeutic approach to diabetes and obesity, which offers reversibility, low risk, low invasiveness and triple benefits including fat mass loss, glucose and lipid metabolism improvement which mechanistically may involve increased villus growth in the upper gut.


Asunto(s)
Glucemia/metabolismo , Duodeno/fisiología , Absorción Intestinal , Síndrome Metabólico/terapia , Prótesis e Implantes , Animales , Ácidos y Sales Biliares/sangre , Composición Corporal , Peso Corporal , Diabetes Mellitus Experimental/terapia , Duodeno/patología , Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Glicerol/sangre , Homeostasis , Íleon/patología , Yeyuno/patología , Masculino , Obesidad/terapia , Distribución Aleatoria , Ratas , Ratas Zucker , Triglicéridos/sangre
3.
Glia ; 62(1): 17-25, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24166765

RESUMEN

The arcuate nucleus (ARC) of the hypothalamus plays a key role in sensing metabolic feedback and regulating energy homeostasis. Recent studies revealed activation of microglia in mice with high-fat diet (HFD)-induced obesity (DIO), suggesting a potential pathophysiological role for inflammatory processes within the hypothalamus. To further investigate the metabolic causes and molecular underpinnings of such glial activation, we analyzed the microglial activity in wild-type (WT), monogenic obese ob/ob (leptin deficient), db/db (leptin-receptor mutation), and Type-4 melanocortin receptor knockout (MC4R KO) mice on either a HFD or on standardized chow (SC) diet. Following HFD exposure, we observed a significant increase in the total number of ARC microglia, immunoreactivity of ionized calcium binding adaptor molecule 1 (iba1-ir), cluster of differentiation 68 (CD68-ir), and ramification of microglial processes. The ob/ob mice had significantly less iba1-ir and ramifications. Leptin replacement rescued these phenomena. The db/db mice had similar iba1-ir comparable with WT mice but had significantly lower CD68-ir and more ramifications than WT mice. After 2 weeks of HFD, ob/ob mice showed an increase of iba1-ir, and db/db mice showed increase of CD68-ir. Obese MC4R KO mice fed a SC diet had comparable iba1-ir and CD68-ir with WT mice but had significantly more ramifications than WT mice. Intriguingly, treatment of DIO mice with glucagon-like peptide-1 receptor agonists reduced microglial activation independent of body weight. Our results show that diet type, adipokines, and gut signals, but not body weight, affect the presence and activity levels of hypothalamic microglia in obesity.


Asunto(s)
Hormonas/farmacología , Microglía/metabolismo , Núcleo Supraóptico/citología , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Peso Corporal/efectos de los fármacos , Peso Corporal/fisiología , Citocininas/metabolismo , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Exenatida , Leptina/deficiencia , Leptina/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Obesidad/inducido químicamente , Obesidad/fisiopatología , Péptidos/farmacología , Receptor de Melanocortina Tipo 4/deficiencia , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/metabolismo , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Transducción de Señal/efectos de los fármacos , Ponzoñas/farmacología
4.
J Pept Sci ; 18(6): 383-93, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22565812

RESUMEN

The identification of leptin as a mediator of body weight regulation provided much initial excitement for the treatment of obesity. Unfortunately, leptin monotherapy is insufficient in reversing obesity in rodents or humans. Recent findings suggest that amylin is able to restore leptin sensitivity and when used in combination with leptin enhances body weight loss in obese rodents and humans. However, as the uniqueness of this combination therapy remains unclear, we assessed whether co-administration of leptin with other weight loss-inducing hormones equally restores leptin responsiveness in diet-induced obese (DIO) mice. Accordingly, we report here the design and characterization of a series of site-specifically enhanced leptin analogs of high potency and sustained action that, when administered in combination with exendin-4 or fibroblast growth factor 21 (FGF21), restores leptin responsiveness in DIO mice after an initial body weight loss of 30%. Using either combination, body weight loss was enhanced compared with either exendin-4 or FGF21 monotherapy, and leptin alone was sufficient to maintain the reduced body weight. In contrast, leptin monotherapy proved ineffective when identical weight loss was induced by caloric restriction alone over a comparable time. Accordingly, we find that a hypothalamic counter-regulatory response to weight loss, assessed using changes in hypothalamic agouti related peptide (AgRP) levels, is triggered by caloric restriction, but blunted by treatment with exendin-4. We conclude that leptin re-sensitization requires pharmacotherapy but does not appear to be restricted to a unique signaling pathway. Our findings provide preclinical evidence that high activity, long-acting leptin analogs are additively efficacious when used in combination with other weight-lowering agents.


Asunto(s)
Dieta/efectos adversos , Factores de Crecimiento de Fibroblastos/farmacología , Leptina/análogos & derivados , Leptina/farmacología , Obesidad/metabolismo , Péptidos/farmacología , Ponzoñas/farmacología , Animales , Peso Corporal , Combinación de Medicamentos , Exenatida , Factores de Crecimiento de Fibroblastos/administración & dosificación , Leptina/administración & dosificación , Leptina/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Modelos Moleculares , Obesidad/inducido químicamente , Obesidad/tratamiento farmacológico , Péptidos/administración & dosificación , Polietilenglicoles/química , Ponzoñas/administración & dosificación
5.
J Neurosci ; 30(17): 6036-47, 2010 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-20427662

RESUMEN

Sepsis describes a complex clinical syndrome that results from an infection, setting off a cascade of systemic inflammatory responses that can lead to multiple organ failure and death. Leptin is a 16 kDa adipokine that, among its multiple known effects, is involved in regulating immune function. Here we demonstrate that leptin deficiency in ob/ob mice leads to higher mortality and more severe organ damage in a standard model of sepsis in mice [cecal ligation and puncture (CLP)]. Moreover, systemic leptin replacement improved the immune response to CLP. Based on the molecular mechanisms of leptin regulation of energy metabolism and reproductive function, we hypothesized that leptin acts in the CNS to efficiently coordinate peripheral immune defense in sepsis. We now report that leptin signaling in the brain increases survival during sepsis in leptin-deficient as well as in wild-type mice and that endogenous CNS leptin action is required for an adequate systemic immune response. These findings reveal the existence of a relevant neuroendocrine control of systemic immune defense and suggest a possible therapeutic potential for leptin analogs in infectious disease.


Asunto(s)
Encéfalo/inmunología , Encéfalo/metabolismo , Leptina/metabolismo , Sepsis/inmunología , Sepsis/metabolismo , Animales , Bacteriemia/inmunología , Bacteriemia/metabolismo , Bacteriemia/mortalidad , Modelos Animales de Enfermedad , Leptina/deficiencia , Leptina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuroinmunomodulación/fisiología , Neutrófilos/metabolismo , Distribución Aleatoria , Receptores de Leptina/deficiencia , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Sepsis/mortalidad
6.
Nat Chem Biol ; 5(10): 749-57, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19597507

RESUMEN

We report the efficacy of a new peptide with agonism at the glucagon and GLP-1 receptors that has potent, sustained satiation-inducing and lipolytic effects. Selective chemical modification to glucagon resulted in a loss of specificity, with minimal change to inherent activity. The structural basis for the co-agonism appears to be a combination of local positional interactions and a change in secondary structure. Two co-agonist peptides differing from each other only in their level of glucagon receptor agonism were studied in rodent obesity models. Administration of PEGylated peptides once per week normalized adiposity and glucose tolerance in diet-induced obese mice. Reduction of body weight was achieved by a loss of body fat resulting from decreased food intake and increased energy expenditure. These preclinical studies indicate that when full GLP-1 agonism is augmented with an appropriate degree of glucagon receptor activation, body fat reduction can be substantially enhanced without any overt adverse effects.


Asunto(s)
Péptido 1 Similar al Glucagón/agonistas , Obesidad/tratamiento farmacológico , Péptidos Cíclicos/uso terapéutico , Polietilenglicoles/química , Receptores de Glucagón/agonistas , Tejido Adiposo/efectos de los fármacos , Secuencia de Aminoácidos , Animales , Peso Corporal/efectos de los fármacos , AMP Cíclico/biosíntesis , Ingestión de Alimentos/efectos de los fármacos , Metabolismo Energético/efectos de los fármacos , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Obesos , Modelos Moleculares , Datos de Secuencia Molecular , Péptidos Cíclicos/química , Péptidos Cíclicos/farmacología , Conformación Proteica
7.
Diabetes ; 67(11): 2157-2166, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30150304

RESUMEN

Glucagon receptor (GCGR) agonists cause hyperglycemia but also weight loss. However, GCG-like peptide 1 receptor (GLP1R)/GCGR mixed agonists do not exhibit the diabetogenic effects often attributed to GCGR activity. Thus, we sought to investigate the effect of glucagon agonism on insulin action and glucose homeostasis. Acute GCGR agonism induced immediate hyperglycemia, followed by improved glucose tolerance and enhanced glucose-stimulated insulin secretion. Moreover, acute GCGR agonism improved insulin tolerance in a dose-dependent manner in both lean and obese mice. Improved insulin tolerance was independent of GLP1R, FGF21, and hepatic glycogenolysis. Moreover, we observed increased glucose infusion rate, disposal, uptake, and suppressed endogenous glucose production during euglycemic clamps. Mice treated with insulin and GCGR agonist had enhanced phosphorylation of hepatic AKT at Ser473; this effect was reproduced in isolated mouse primary hepatocytes and resulted in increased AKT kinase activity. These data reveal that GCGR agonism enhances glucose tolerance, in part, by augmenting insulin action, with implications for the use of GCGR agonism in therapeutic strategies for diabetes.


Asunto(s)
Glucosa/metabolismo , Insulina/metabolismo , Hígado/metabolismo , Receptores de Glucagón/metabolismo , Animales , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Prueba de Tolerancia a la Glucosa , Insulina/farmacología , Resistencia a la Insulina/fisiología , Hígado/efectos de los fármacos , Ratones , Ratones Noqueados , Obesidad/metabolismo , Péptidos/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Glucagón/agonistas
8.
Cell Metab ; 21(6): 877-82, 2015 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-25980347

RESUMEN

Obesity is characterized by hyperleptinemia and decreased response to exogenous leptin. This has been widely attributed to the development of leptin resistance, a state of impaired leptin signaling proposed to contribute to the development and persistence of obesity. To directly determine endogenous leptin activity in obesity, we treated lean and obese mice with a leptin receptor antagonist. The antagonist increased feeding and body weight (BW) in lean mice, but not in obese models of leptin, leptin receptor, or melanocortin-4 receptor deficiency. In contrast, the antagonist increased feeding and BW comparably in lean and diet-induced obese (DIO) mice, an increase associated with decreased hypothalamic expression of Socs3, a primary target of leptin. These findings demonstrate that hyperleptinemic DIO mice retain leptin suppression of feeding comparable to lean mice and counter the view that resistance to endogenous leptin contributes to the persistence of DIO in mice.


Asunto(s)
Dieta/efectos adversos , Leptina/metabolismo , Obesidad/metabolismo , Animales , Peso Corporal/genética , Hiperlipidemias/inducido químicamente , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Leptina/genética , Ratones , Ratones Noqueados , Ratones Obesos , Obesidad/inducido químicamente , Obesidad/genética , Receptor de Melanocortina Tipo 4/genética , Receptor de Melanocortina Tipo 4/metabolismo , Receptores de Leptina/antagonistas & inhibidores , Receptores de Leptina/genética , Receptores de Leptina/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
9.
Endocrinology ; 156(5): 1685-91, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25730108

RESUMEN

The melanocortin system directs diverse physiological functions from coat color to body weight homoeostasis. A commonality among melanocortin-mediated processes is that many animals modulate similar processes on a circannual basis in response to longer, summer days, suggesting an underlying link between circadian biology and the melanocortin system. Despite key neuroanatomical substrates shared by both circadian and melanocortin-signaling pathways, little is known about the relationship between the two. Here we identify a link between circadian disruption and the control of glucose homeostasis mediated through the melanocortin-4 receptor (Mc4r). Mc4r-deficient mice exhibit exaggerated circadian fluctuations in baseline blood glucose and glucose tolerance. Interestingly, exposure to lighting conditions that disrupt circadian rhythms improve their glucose tolerance. This improvement occurs through an increase in glucose clearance by skeletal muscle and is food intake and body weight independent. Restoring Mc4r expression to the paraventricular nucleus prevents the improvement in glucose tolerance, supporting a role for the paraventricular nucleus in the integration of circadian light cues and metabolism. Altogether these data suggest that Mc4r signaling plays a protective role in minimizing glucose fluctuations due to circadian rhythms and environmental light cues and demonstrate a previously undiscovered connection between circadian biology and glucose metabolism mediated through the melanocortin system.


Asunto(s)
Glucemia/metabolismo , Ritmo Circadiano/genética , Iluminación , Músculo Esquelético/metabolismo , Núcleo Hipotalámico Paraventricular/metabolismo , Receptor de Melanocortina Tipo 4/genética , Animales , Señales (Psicología) , Técnica de Clampeo de la Glucosa , Prueba de Tolerancia a la Glucosa , Ratones , Ratones Noqueados , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal/genética
10.
Nat Med ; 21(1): 27-36, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25485909

RESUMEN

We report the discovery of a new monomeric peptide that reduces body weight and diabetic complications in rodent models of obesity by acting as an agonist at three key metabolically-related peptide hormone receptors: glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP) and glucagon receptors. This triple agonist demonstrates supraphysiological potency and equally aligned constituent activities at each receptor, all without cross-reactivity at other related receptors. Such balanced unimolecular triple agonism proved superior to any existing dual coagonists and best-in-class monoagonists to reduce body weight, enhance glycemic control and reverse hepatic steatosis in relevant rodent models. Various loss-of-function models, including genetic knockout, pharmacological blockade and selective chemical knockout, confirmed contributions of each constituent activity in vivo. We demonstrate that these individual constituent activities harmonize to govern the overall metabolic efficacy, which predominantly results from synergistic glucagon action to increase energy expenditure, GLP-1 action to reduce caloric intake and improve glucose control, and GIP action to potentiate the incretin effect and buffer against the diabetogenic effect of inherent glucagon activity. These preclinical studies suggest that, so far, this unimolecular, polypharmaceutical strategy has potential to be the most effective pharmacological approach to reversing obesity and related metabolic disorders.


Asunto(s)
Complicaciones de la Diabetes/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Obesidad/metabolismo , Péptidos/administración & dosificación , Animales , Glucemia/efectos de los fármacos , Peso Corporal/genética , Complicaciones de la Diabetes/tratamiento farmacológico , Complicaciones de la Diabetes/genética , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/genética , Péptido 1 Similar al Glucagón/agonistas , Péptido 1 Similar al Glucagón/metabolismo , Células HEK293 , Humanos , Insulina/biosíntesis , Insulina/metabolismo , Ratones , Obesidad/tratamiento farmacológico , Obesidad/genética , Péptidos/síntesis química , Péptidos/metabolismo , Ratas , Receptores de la Hormona Gastrointestinal/agonistas , Receptores de la Hormona Gastrointestinal/metabolismo , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Roedores
11.
Diabetes ; 63(1): 122-31, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24062249

RESUMEN

Growth hormone secretagogue receptors (GHSRs) in the central nervous system (CNS) mediate hyperphagia and adiposity induced by acyl ghrelin (AG). Evidence suggests that des-AG (dAG) has biological activity through GHSR-independent mechanisms. We combined in vitro and in vivo approaches to test possible GHSR-mediated biological activity of dAG. Both AG (100 nmol/L) and dAG (100 nmol/L) significantly increased inositol triphosphate formation in human embryonic kidney-293 cells transfected with human GHSR. As expected, intracerebroventricular infusion of AG in mice increased fat mass (FM), in comparison with the saline-infused controls. Intracerebroventricular dAG also increased FM at the highest dose tested (5 nmol/day). Chronic intracerebroventricular infusion of AG or dAG increased glucose-stimulated insulin secretion (GSIS). Subcutaneously infused AG regulated FM and GSIS in comparison with saline-infused control mice, whereas dAG failed to regulate these parameters even with doses that were efficacious when delivered intracerebroventricularly. Furthermore, intracerebroventricular dAG failed to regulate FM and induce hyperinsulinemia in GHSR-deficient (Ghsr(-/-)) mice. In addition, a hyperinsulinemic-euglycemic clamp suggests that intracerebroventricular dAG impairs glucose clearance without affecting endogenous glucose production. Together, these data demonstrate that dAG is an agonist of GHSR and regulates body adiposity and peripheral glucose metabolism through a CNS GHSR-dependent mechanism.


Asunto(s)
Adiposidad/fisiología , Ghrelina/farmacología , Glucosa/metabolismo , Receptores de Ghrelina/metabolismo , Adiposidad/efectos de los fármacos , Animales , Sistema Nervioso Central/metabolismo , Ghrelina/administración & dosificación , Células HEK293 , Humanos , Infusiones Intraventriculares , Ratones
12.
Diabetes ; 63(2): 505-13, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24186863

RESUMEN

Several bariatric operations are currently used to treat obesity and obesity-related comorbidities. These vary in efficacy, but most are more effective than current pharmaceutical treatments. Roux-en-Y gastric bypass (RYGB) produces substantial body weight (BW) loss and enhanced glucose tolerance, and is associated with increased secretion of the gut hormone glucagon-like peptide 1 (GLP-1). Given the success of GLP-1-based agents in lowering blood glucose levels and BW, we hypothesized that an individual sensitivity to GLP-1 receptor agonism could predict metabolic benefits of surgeries associated with increased GLP-1 secretion. One hundred ninety-seven high-fat diet-induced obese male Long-Evans rats were monitored for BW loss during exendin-4 (Ex4) administration. Stable populations of responders and nonresponders were identified based on Ex4-induced BW loss and GLP-1-induced improvements in glucose tolerance. Subpopulations of Ex4 extreme responders and nonresponders underwent RYGB surgery. After RYGB, responders and nonresponders showed similar BW loss compared with sham, but nonresponders retained impaired glucose tolerance. These data indicate that the GLP-1 response tests may predict some but not all of the improvements observed after RYGB. These findings present an opportunity to optimize the use of bariatric surgery based on an improved understanding of GLP-1 biology and suggest an opportunity for a more personalized therapeutic approach to the metabolic syndrome.


Asunto(s)
Derivación Gástrica , Prueba de Tolerancia a la Glucosa , Receptores de Glucagón/metabolismo , Animales , Grasas de la Dieta/efectos adversos , Ingestión de Alimentos , Exenatida , Regulación de la Expresión Génica/fisiología , Receptor del Péptido 1 Similar al Glucagón , Masculino , Obesidad , Péptidos/farmacología , Ratas , Ratas Long-Evans , Receptores de Glucagón/agonistas , Receptores de Glucagón/genética , Ponzoñas/farmacología , Pérdida de Peso
13.
Mol Metab ; 2(4): 468-79, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24327962

RESUMEN

Medicines that decrease body weight and restore nutrient tolerance could improve human diabetes and obesity treatment outcomes. We developed lipid-acylated glucagon analogs that are co-agonists for the glucagon and glucagon-like peptide 1 receptors, and stimulate weight loss and plasma glucose lowering in pre-diabetic obese mice. Our studies identified lipid acylation (lipidation) can increase and balance in vitro potencies of select glucagon analogs for the two aforementioned receptors in a lipidation site-dependent manner. A general capacity for lipidation to enhance the secondary structure of glucagon analogs was recognized, and the energetics of this effect quantified. The molecular structure of a lipid-acylated glucagon analog in water was also characterized. These results support that lipidation can modify biological activity through thermodynamically-favorable intramolecular interactions which stabilize structure. This establishes use of lipidation to achieve specific pharmacology and implicates similar endogenous post-translational modifications as physiological tools capable of refining biological action in means previously underappreciated.

14.
Diabetes ; 62(9): 3261-7, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23775764

RESUMEN

Bariatric procedures vary in efficacy, but overall are more effective than behavioral and pharmaceutical treatment. Roux-en-Y gastric bypass causes increased secretion of glucagon-like peptide 1 (GLP-1) and reduces body weight (BW) more than adjustable gastric banding (AGB), which does not trigger increased GLP-1 secretion. Since GLP-1-based drugs consistently reduce BW, we hypothesized that GLP-1 receptor (GLP-1R) agonists would augment the effects of AGB. Male Long-Evans rats with diet-induced obesity received AGB implantation or sham surgery. GLP-1R agonism, cannabinoid receptor-1 (CB1-R) antagonism, or vehicle was combined with inflation to evaluate interaction between AGB and pharmacological treatments. GLP1-R agonism reduced BW in both sham and AGB rats (left uninflated) compared with vehicle-treated animals. Subsequent band inflation was ineffective in vehicle-treated rats but enhanced weight loss stimulated by GLP1-R agonism. In contrast, there was no additional BW loss when CB1-R antagonism was given with AGB. We found band inflation to trigger neural activation in areas of the nucleus of the solitary tract known to be targeted by GLP-1R agonism, offering a potential mechanism for the interaction. These data show that GLP-1R agonism, but not CB1-R antagonism, improves weight loss achieved by AGB and suggest an opportunity to optimize bariatric surgery with adjunctive pharmacotherapy.


Asunto(s)
Obesidad/tratamiento farmacológico , Obesidad/cirugía , Receptores de Glucagón/agonistas , Animales , Composición Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Exenatida , Derivación Gástrica , Gastroplastia , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón , Inmunohistoquímica , Masculino , Obesidad/etiología , Obesidad/metabolismo , Péptidos/uso terapéutico , Ratas , Ratas Long-Evans , Receptores de Cannabinoides/metabolismo , Ponzoñas/uso terapéutico
15.
Sci Transl Med ; 5(209): 209ra151, 2013 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-24174327

RESUMEN

We report the discovery and translational therapeutic efficacy of a peptide with potent, balanced co-agonism at both of the receptors for the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). This unimolecular dual incretin is derived from an intermixed sequence of GLP-1 and GIP, and demonstrated enhanced antihyperglycemic and insulinotropic efficacy relative to selective GLP-1 agonists. Notably, this superior efficacy translated across rodent models of obesity and diabetes, including db/db mice and ZDF rats, to primates (cynomolgus monkeys and humans). Furthermore, this co-agonist exhibited synergism in reducing fat mass in obese rodents, whereas a selective GIP agonist demonstrated negligible weight-lowering efficacy. The unimolecular dual incretins corrected two causal mechanisms of diabesity, adiposity-induced insulin resistance and pancreatic insulin deficiency, more effectively than did selective mono-agonists. The duration of action of the unimolecular dual incretins was refined through site-specific lipidation or PEGylation to support less frequent administration. These peptides provide comparable pharmacology to the native peptides and enhanced efficacy relative to similarly modified selective GLP-1 agonists. The pharmacokinetic enhancement lessened peak drug exposure and, in combination with less dependence on GLP-1-mediated pharmacology, avoided the adverse gastrointestinal effects that typify selective GLP-1-based agonists. This discovery and validation of a balanced and high-potency dual incretin agonist enables a more physiological approach to management of diseases associated with impaired glucose tolerance.


Asunto(s)
Haplorrinos/metabolismo , Incretinas/farmacología , Roedores/metabolismo , Acilación/efectos de los fármacos , Adolescente , Adulto , Anciano , Animales , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Exenatida , Femenino , Polipéptido Inhibidor Gástrico/administración & dosificación , Polipéptido Inhibidor Gástrico/farmacología , Péptido 1 Similar al Glucagón/administración & dosificación , Péptido 1 Similar al Glucagón/análogos & derivados , Péptido 1 Similar al Glucagón/farmacología , Receptor del Péptido 1 Similar al Glucagón , Prueba de Tolerancia a la Glucosa , Humanos , Hiperglucemia/tratamiento farmacológico , Incretinas/administración & dosificación , Incretinas/uso terapéutico , Insulina/metabolismo , Liraglutida , Masculino , Ratones , Persona de Mediana Edad , Péptidos/farmacología , Ratas , Receptores de la Hormona Gastrointestinal , Receptores de Glucagón/agonistas , Receptores de Glucagón/metabolismo , Resultado del Tratamiento , Ponzoñas/farmacología , Pérdida de Peso/efectos de los fármacos , Adulto Joven
16.
Diabetes ; 62(5): 1453-63, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23305646

RESUMEN

Glucagon, an essential regulator of glucose homeostasis, also modulates lipid metabolism and promotes weight loss, as reflected by the wasting observed in glucagonoma patients. Recently, coagonist peptides that include glucagon agonism have emerged as promising therapeutic candidates for the treatment of obesity and diabetes. We developed a novel stable and soluble glucagon receptor (GcgR) agonist, which allowed for in vivo dissection of glucagon action. As expected, chronic GcgR agonism in mice resulted in hyperglycemia and lower body fat and plasma cholesterol. Notably, GcgR activation also raised hepatic expression and circulating levels of fibroblast growth factor 21 (FGF21). This effect was retained in isolated primary hepatocytes from wild-type (WT) mice, but not GcgR knockout mice. We confirmed this link in healthy human volunteers, where injection of natural glucagon increased plasma FGF21 within hours. Functional relevance was evidenced in mice with genetic deletion of FGF21, where GcgR activation failed to induce the body weight loss and lipid metabolism changes observed in WT mice. Taken together, these data reveal for the first time that glucagon controls glucose, energy, and lipid metabolism at least in part via FGF21-dependent pathways.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucagón/metabolismo , Hepatocitos/metabolismo , Receptores de Glucagón/metabolismo , Adulto , Animales , Fármacos Antiobesidad/síntesis química , Fármacos Antiobesidad/farmacocinética , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Células Cultivadas , Estudios Cruzados , Diabetes Mellitus Tipo 2/sangre , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Método Doble Ciego , Femenino , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/genética , Glucagón/agonistas , Glucagón/farmacología , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/patología , Humanos , Hipoglucemiantes/síntesis química , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina , Masculino , Ratones , Ratones Noqueados , Ratones Mutantes , Terapia Molecular Dirigida , Obesidad/sangre , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , Péptidos/síntesis química , Péptidos/farmacocinética , Péptidos/fisiología , Péptidos/uso terapéutico , Ratas , Receptores de Glucagón/agonistas , Receptores de Glucagón/genética , Proteínas Recombinantes/agonistas , Proteínas Recombinantes/metabolismo
17.
J Clin Invest ; 123(1): 469-78, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23257354

RESUMEN

The scaffold protein p62 (sequestosome 1; SQSTM1) is an emerging key molecular link among the metabolic, immune, and proliferative processes of the cell. Here, we report that adipocyte-specific, but not CNS-, liver-, muscle-, or myeloid-specific p62-deficient mice are obese and exhibit a decreased metabolic rate caused by impaired nonshivering thermogenesis. Our results show that p62 regulates energy metabolism via control of mitochondrial function in brown adipose tissue (BAT). Accordingly, adipocyte-specific p62 deficiency led to impaired mitochondrial function, causing BAT to become unresponsive to ß-adrenergic stimuli. Ablation of p62 leads to decreased activation of p38 targets, affecting signaling molecules that control mitochondrial function, such as ATF2, CREB, PGC1α, DIO2, NRF1, CYTC, COX2, ATP5ß, and UCP1. p62 ablation in HIB1B and BAT primary cells demonstrated that p62 controls thermogenesis in a cell-autonomous manner, independently of brown adipocyte development or differentiation. Together, our data identify p62 as a novel regulator of mitochondrial function and brown fat thermogenesis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/metabolismo , Proteínas de Choque Térmico/metabolismo , Mitocondrias/metabolismo , Termogénesis/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Adipocitos Marrones/citología , Tejido Adiposo Pardo/citología , Animales , Células Cultivadas , Proteínas de Choque Térmico/genética , Sistema de Señalización de MAP Quinasas/fisiología , Ratones , Ratones Noqueados , Mitocondrias/genética , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Especificidad de Órganos/genética , Proteína Sequestosoma-1 , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
18.
Nat Commun ; 4: 1968, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23744028

RESUMEN

The G protein-coupled receptor 83 (Gpr83) is widely expressed in brain regions regulating energy metabolism. Here we report that hypothalamic expression of Gpr83 is regulated in response to nutrient availability and is decreased in obese mice compared with lean mice. In the arcuate nucleus, Gpr83 colocalizes with the ghrelin receptor (Ghsr1a) and the agouti-related protein. In vitro analyses show heterodimerization of Gpr83 with Ghsr1a diminishes activation of Ghsr1a by acyl-ghrelin. The orexigenic and adipogenic effect of ghrelin is accordingly potentiated in Gpr83-deficient mice. Interestingly, Gpr83 knock-out mice have normal body weight and glucose tolerance when fed a regular chow diet, but are protected from obesity and glucose intolerance when challenged with a high-fat diet, despite hyperphagia and increased hypothalamic expression of agouti-related protein, Npy, Hcrt and Ghsr1a. Together, our data suggest that Gpr83 modulates ghrelin action but also indicate that Gpr83 regulates systemic metabolism through other ghrelin-independent pathways.


Asunto(s)
Metabolismo Energético , Ghrelina/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteína Relacionada con Agouti/metabolismo , Animales , Núcleo Arqueado del Hipotálamo/efectos de los fármacos , Núcleo Arqueado del Hipotálamo/metabolismo , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Dieta Alta en Grasa , Metabolismo Energético/efectos de los fármacos , Conducta Alimentaria/efectos de los fármacos , Perfilación de la Expresión Génica , Ghrelina/administración & dosificación , Ghrelina/farmacología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Fenotipo , Multimerización de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Ratas , Receptor de Melanocortina Tipo 3/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores de Ghrelina/metabolismo , Transducción de Señal/efectos de los fármacos
19.
Diabetes ; 61(11): 2734-42, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22787140

RESUMEN

Although obesity rates are rapidly rising, caloric restriction remains one of the few safe therapies. Here we tested the hypothesis that obesity-associated disorders are caused by increased adipose tissue as opposed to excess dietary lipids. Fat mass (FM) of lean C57B6 mice fed a high-fat diet (HFD; FMC mice) was "clamped" to match the FM of mice maintained on a low-fat diet (standard diet [SD] mice). FMC mice displayed improved glucose and insulin tolerance as compared with ad libitum HFD mice (P < 0.001) or SD mice (P < 0.05). These improvements were associated with fewer signs of inflammation, consistent with the less-impaired metabolism. In follow-up studies, diet-induced obese mice were food restricted for 5 weeks to achieve FM levels identical with those of age-matched SD mice. Previously, obese mice exhibited improved glucose and insulin tolerance but showed markedly increased fasting-induced hyperphagia (P < 0.001). When mice were given ad libitum access to the HFD, the hyperphagia of these mice led to accelerated body weight gain as compared with otherwise matched controls without a history of obesity. These results suggest that although caloric restriction on a HFD provides metabolic benefits, maintaining those benefits may require lifelong continuation, at least in individuals with a history of obesity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Restricción Calórica/efectos adversos , Metabolismo Energético , Adiposidad , Animales , Dieta Alta en Grasa/efectos adversos , Dieta Reductora/efectos adversos , Regulación de la Expresión Génica , Intolerancia a la Glucosa/sangre , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/inmunología , Intolerancia a la Glucosa/metabolismo , Hiperfagia/etiología , Hipotálamo/metabolismo , Mediadores de Inflamación/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/dietoterapia , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/prevención & control , Análisis de Secuencia por Matrices de Oligonucleótidos , Especificidad de Órganos , Distribución Aleatoria , Prevención Secundaria , Aumento de Peso
20.
Endocrinology ; 153(10): 4687-95, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22865372

RESUMEN

Ghrelin is a gastrointestinal polypeptide that acts through the ghrelin receptor (GHSR) to promote food intake and increase adiposity. Activation of GHSR requires the presence of a fatty-acid (FA) side chain on amino acid residue serine 3 of the ghrelin molecule. However, little is known about the role that the type of FA used for acylation plays in the biological action of ghrelin. We therefore evaluated a series of differentially acylated peptides to determine whether alterations in length or stability of the FA side chain have an impact on the ability of ghrelin to activate GHSR in vitro or to differentially alter food intake, body weight, and body composition in vivo. Fatty acids principally available in the diet (such as palmitate C16) and therefore representing potential substrates for the ghrelin-activating enzyme ghrelin O-acyltransferase (GOAT) were used for dose-, time-, and administration/route-dependent effects of ghrelin on food intake, body weight, and body composition in rats and mice. Our data demonstrate that altering the length of the FA side chain of ghrelin results in the differential activation of GHSR. Additionally, we found that acylation of ghrelin with a long-chain FA (C16) delays the acute central stimulation of food intake. Lastly, we found that, depending on acylation length, systemic and central chronic actions of ghrelin on adiposity can be enhanced or reduced. Together our data suggest that modification of the FA side-chain length can be a novel approach to modulate the efficacy of pharmacologically administered ghrelin.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Ghrelina/metabolismo , Homeostasis/efectos de los fármacos , Receptores de Ghrelina/genética , Acilación , Animales , Composición Corporal/efectos de los fármacos , Peso Corporal/efectos de los fármacos , Ingestión de Alimentos/efectos de los fármacos , Ghrelina/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/farmacología , Ratas , Ratas Long-Evans , Receptores de Ghrelina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA