Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Diabetes ; 67(11): 2157-2166, 2018 11.
Article in English | MEDLINE | ID: mdl-30150304

ABSTRACT

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.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Liver/metabolism , Receptors, Glucagon/metabolism , Animals , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test , Insulin/pharmacology , Insulin Resistance/physiology , Liver/drug effects , Mice , Mice, Knockout , Obesity/metabolism , Peptides/pharmacology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Glucagon/agonists
2.
Diabetologia ; 58(9): 2124-32, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26049402

ABSTRACT

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.


Subject(s)
Adipose Tissue, Brown/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Body Composition , Calorimetry, Indirect , Diet , Diet, High-Fat , Eating , Energy Metabolism/physiology , Liraglutide/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Norepinephrine/chemistry , Oxygen Consumption , Phenotype , Signal Transduction , Temperature , Thermogenesis
3.
Cell Metab ; 21(6): 877-82, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-25980347

ABSTRACT

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.


Subject(s)
Diet/adverse effects , Leptin/metabolism , Obesity/metabolism , Animals , Body Weight/genetics , Hyperlipidemias/chemically induced , Hyperlipidemias/genetics , Hyperlipidemias/metabolism , Leptin/genetics , Mice , Mice, Knockout , Mice, Obese , Obesity/chemically induced , Obesity/genetics , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Leptin/antagonists & inhibitors , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism
4.
Endocrinology ; 156(5): 1685-91, 2015 May.
Article in English | MEDLINE | ID: mdl-25730108

ABSTRACT

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.


Subject(s)
Blood Glucose/metabolism , Circadian Rhythm/genetics , Lighting , Muscle, Skeletal/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 4/genetics , Animals , Cues , Glucose Clamp Technique , Glucose Tolerance Test , Mice , Mice, Knockout , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/genetics
5.
Nat Med ; 21(1): 27-36, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485909

ABSTRACT

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.


Subject(s)
Diabetes Complications/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Peptides/administration & dosage , Animals , Blood Glucose/drug effects , Body Weight/genetics , Diabetes Complications/drug therapy , Diabetes Complications/genetics , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/metabolism , HEK293 Cells , Humans , Insulin/biosynthesis , Insulin/metabolism , Mice , Obesity/drug therapy , Obesity/genetics , Peptides/chemical synthesis , Peptides/metabolism , Rats , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Rodentia
6.
Gut ; 63(8): 1238-46, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24107591

ABSTRACT

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.


Subject(s)
Blood Glucose/metabolism , Duodenum/physiology , Intestinal Absorption , Metabolic Syndrome/therapy , Prostheses and Implants , Animals , Bile Acids and Salts/blood , Body Composition , Body Weight , Diabetes Mellitus, Experimental/therapy , Duodenum/pathology , Glucagon-Like Peptide 1/metabolism , Glucose Tolerance Test , Glycerol/blood , Homeostasis , Ileum/pathology , Jejunum/pathology , Male , Obesity/therapy , Random Allocation , Rats , Rats, Zucker , Triglycerides/blood
7.
Diabetes ; 63(2): 505-13, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24186863

ABSTRACT

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.


Subject(s)
Gastric Bypass , Glucose Tolerance Test , Receptors, Glucagon/metabolism , Animals , Dietary Fats/adverse effects , Eating , Exenatide , Gene Expression Regulation/physiology , Glucagon-Like Peptide-1 Receptor , Male , Obesity , Peptides/pharmacology , Rats , Rats, Long-Evans , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Venoms/pharmacology , Weight Loss
8.
Diabetes ; 63(1): 122-31, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24062249

ABSTRACT

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.


Subject(s)
Adiposity/physiology , Ghrelin/pharmacology , Glucose/metabolism , Receptors, Ghrelin/metabolism , Adiposity/drug effects , Animals , Central Nervous System/metabolism , Ghrelin/administration & dosage , HEK293 Cells , Humans , Infusions, Intraventricular , Mice
9.
Glia ; 62(1): 17-25, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24166765

ABSTRACT

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.


Subject(s)
Hormones/pharmacology , Microglia/metabolism , Supraoptic Nucleus/cytology , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Body Weight/drug effects , Body Weight/physiology , Cytokinins/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Exenatide , Leptin/deficiency , Leptin/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Obesity/chemically induced , Obesity/physiopathology , Peptides/pharmacology , Receptor, Melanocortin, Type 4/deficiency , Receptors, Interleukin-8A/genetics , Receptors, Interleukin-8A/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/genetics , Signal Transduction/drug effects , Venoms/pharmacology
10.
Mol Metab ; 2(4): 468-79, 2013.
Article in English | MEDLINE | ID: mdl-24327962

ABSTRACT

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.

11.
Sci Transl Med ; 5(209): 209ra151, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24174327

ABSTRACT

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.


Subject(s)
Haplorhini/metabolism , Incretins/pharmacology , Rodentia/metabolism , Acylation/drug effects , Adolescent , Adult , Aged , Animals , Diabetes Mellitus, Type 2/drug therapy , Exenatide , Female , Gastric Inhibitory Polypeptide/administration & dosage , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Humans , Hyperglycemia/drug therapy , Incretins/administration & dosage , Incretins/therapeutic use , Insulin/metabolism , Liraglutide , Male , Mice , Middle Aged , Peptides/pharmacology , Rats , Receptors, Gastrointestinal Hormone , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Treatment Outcome , Venoms/pharmacology , Weight Loss/drug effects , Young Adult
12.
Diabetes ; 62(9): 3261-7, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23775764

ABSTRACT

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.


Subject(s)
Obesity/drug therapy , Obesity/surgery , Receptors, Glucagon/agonists , Animals , Body Composition/drug effects , Eating/drug effects , Exenatide , Gastric Bypass , Gastroplasty , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Immunohistochemistry , Male , Obesity/etiology , Obesity/metabolism , Peptides/therapeutic use , Rats , Rats, Long-Evans , Receptors, Cannabinoid/metabolism , Venoms/therapeutic use
13.
Nat Commun ; 4: 1968, 2013.
Article in English | MEDLINE | ID: mdl-23744028

ABSTRACT

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.


Subject(s)
Energy Metabolism , Ghrelin/metabolism , Receptors, G-Protein-Coupled/metabolism , Agouti-Related Protein/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Body Composition/drug effects , Body Weight/drug effects , Diet, High-Fat , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Gene Expression Profiling , Ghrelin/administration & dosage , Ghrelin/pharmacology , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Phenotype , Protein Multimerization/drug effects , Protein Transport/drug effects , Rats , Receptor, Melanocortin, Type 3/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin/metabolism , Signal Transduction/drug effects
14.
Diabetes ; 62(5): 1453-63, 2013 May.
Article in English | MEDLINE | ID: mdl-23305646

ABSTRACT

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.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucagon/metabolism , Hepatocytes/metabolism , Receptors, Glucagon/metabolism , Adult , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Cells, Cultured , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Double-Blind Method , Female , Fibroblast Growth Factors/blood , Fibroblast Growth Factors/genetics , Glucagon/agonists , Glucagon/pharmacology , HEK293 Cells , Hepatocytes/drug effects , Hepatocytes/pathology , Humans , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Male , Mice , Mice, Knockout , Mice, Mutant Strains , Molecular Targeted Therapy , Obesity/blood , Obesity/drug therapy , Obesity/metabolism , Peptides/chemical synthesis , Peptides/pharmacokinetics , Peptides/physiology , Peptides/therapeutic use , Rats , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Recombinant Proteins/agonists , Recombinant Proteins/metabolism
15.
J Clin Invest ; 123(1): 469-78, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23257354

ABSTRACT

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.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Heat-Shock Proteins/metabolism , Mitochondria/metabolism , Thermogenesis/physiology , Adaptor Proteins, Signal Transducing/genetics , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Animals , Cells, Cultured , Heat-Shock Proteins/genetics , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mitochondria/genetics , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Organ Specificity/genetics , Sequestosome-1 Protein , Transcription Factors/genetics , Transcription Factors/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Nat Med ; 18(12): 1847-56, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23142820

ABSTRACT

We report the development of a new combinatorial approach that allows for peptide-mediated selective tissue targeting of nuclear hormone pharmacology while eliminating adverse effects in other tissues. Specifically, we report the development of a glucagon-like peptide-1 (GLP-1)-estrogen conjugate that has superior sex-independent efficacy over either of the individual hormones alone to correct obesity, hyperglycemia and dyslipidemia in mice. The therapeutic benefits are driven by pleiotropic dual hormone action to improve energy, glucose and lipid metabolism, as shown by loss-of-function models and genetic action profiling. Notably, the peptide-based targeting strategy also prevents hallmark side effects of estrogen in male and female mice, such as reproductive endocrine toxicity and oncogenicity. Collectively, selective activation of estrogen receptors in GLP-1-targeted tissues produces unprecedented efficacy to enhance the metabolic benefits of GLP-1 agonism. This example of targeting the metabolic syndrome represents the discovery of a new class of therapeutics that enables synergistic co-agonism through peptide-based selective delivery of small molecules. Although our observations with the GLP-1-estrogen conjugate justify translational studies for diabetes and obesity, the multitude of other possible combinations of peptides and small molecules may offer equal promise for other diseases.


Subject(s)
Estrogens/pharmacology , Glucagon-Like Peptide 1/pharmacology , Metabolic Syndrome/drug therapy , Receptors, Estrogen/metabolism , Analysis of Variance , Animals , Binding, Competitive , Body Composition/physiology , Chromatography, High Pressure Liquid , Drug Combinations , Drug Discovery , Estrogens/metabolism , Estrogens/therapeutic use , Female , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/therapeutic use , Glucose Tolerance Test , Humans , MCF-7 Cells , Magnetic Resonance Imaging , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Estrogen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
17.
Endocrinology ; 153(10): 4687-95, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22865372

ABSTRACT

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.


Subject(s)
Energy Metabolism/drug effects , Ghrelin/metabolism , Homeostasis/drug effects , Receptors, Ghrelin/genetics , Acylation , Animals , Body Composition/drug effects , Body Weight/drug effects , Eating/drug effects , Ghrelin/pharmacology , Male , Mice , Mice, Inbred C57BL , Protein Isoforms/metabolism , Protein Isoforms/pharmacology , Rats , Rats, Long-Evans , Receptors, Ghrelin/metabolism
18.
Diabetes ; 61(11): 2734-42, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22787140

ABSTRACT

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.


Subject(s)
Adipose Tissue, White/metabolism , Caloric Restriction/adverse effects , Energy Metabolism , Adiposity , Animals , Diet, High-Fat/adverse effects , Diet, Reducing/adverse effects , Gene Expression Regulation , Glucose Intolerance/blood , Glucose Intolerance/etiology , Glucose Intolerance/immunology , Glucose Intolerance/metabolism , Hyperphagia/etiology , Hypothalamus/metabolism , Inflammation Mediators/metabolism , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Obesity/diet therapy , Obesity/immunology , Obesity/metabolism , Obesity/prevention & control , Oligonucleotide Array Sequence Analysis , Organ Specificity , Random Allocation , Secondary Prevention , Weight Gain
19.
J Pept Sci ; 18(6): 383-93, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22565812

ABSTRACT

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.


Subject(s)
Diet/adverse effects , Fibroblast Growth Factors/pharmacology , Leptin/analogs & derivatives , Leptin/pharmacology , Obesity/metabolism , Peptides/pharmacology , Venoms/pharmacology , Animals , Body Weight , Drug Combinations , Exenatide , Fibroblast Growth Factors/administration & dosage , Leptin/administration & dosage , Leptin/therapeutic use , Mice , Mice, Inbred C57BL , Mice, Obese , Models, Molecular , Obesity/chemically induced , Obesity/drug therapy , Peptides/administration & dosage , Polyethylene Glycols/chemistry , Venoms/administration & dosage
20.
PLoS One ; 7(2): e32100, 2012.
Article in English | MEDLINE | ID: mdl-22363801

ABSTRACT

OBJECTIVE: Ghrelin acylation by ghrelin O-acyltransferase (GOAT) has recently been reported to be essential for the prevention of hypoglycemia during prolonged negative energy balance. Using a unique set of four different genetic loss-of-function models for the GOAT/ghrelin/growth hormone secretagogue receptor (GHSR) system, we thoroughly tested the hypothesis that lack-of-ghrelin activation or signaling would lead to hypoglycemia during caloric deprivation. METHODOLOGY: Male and female knockout (KO) mice for GOAT, ghrelin, GHSR, or both ghrelin and GHSR (dKO) were subjected to prolonged calorie restriction (40% of ad libitum chow intake). Body weight, fat mass, and glucose levels were recorded daily and compared to wildtype (WT) controls. Forty-eight hour blood glucose profiles were generated for each individual mouse when 2% or less body fat mass was reached. Blood samples were obtained for analysis of circulating levels of acyl- and desacyl-ghrelin, IGF-1, and insulin. PRINCIPAL FINDINGS: Chronic calorie restriction progressively decreased body weight and body fat mass in all mice regardless of genotype. When fat mass was depleted to 2% or less of body weight for 2 consecutive days, random hypoglycemic events occurred in some mice across all genotypes. There was no increase in the incidence of hypoglycemia in any of the four loss-of-function models for ghrelin signaling including GOAT KO mice. Furthermore, no differences in insulin or IGF-1 levels were observed between genotypes. CONCLUSION: The endogenous GOAT-ghrelin-GHSR system is not essential for the maintenance of euglycemia during prolonged calorie restriction.


Subject(s)
Acyltransferases/metabolism , Caloric Restriction , Ghrelin/metabolism , Hypoglycemia/prevention & control , Adiposity , Animals , Blood Glucose/metabolism , Body Weight , Female , Genotype , Ghrelin/blood , Hypoglycemia/blood , Insulin/blood , Insulin-Like Growth Factor I/metabolism , Male , Membrane Proteins , Mice , Mice, Knockout , Models, Animal
SELECTION OF CITATIONS
SEARCH DETAIL
...