Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
Mol Metab ; 2(3): 171-83, 2013.
Article in English | MEDLINE | ID: mdl-24049732

ABSTRACT

Alterations in the gut microbiota have been proposed to modify the development and maintenance of obesity and its sequelae. Definition of underlying mechanisms has lagged, although the ability of commensal gut microbes to drive pathways involved in inflammation and metabolism has generated compelling, testable hypotheses. We studied C57BL/6 mice from two vendors that differ in their obesogenic response and in their colonization by specific members of the gut microbiota having well-described roles in regulating gut immune responses. We confirmed the presence of robust differences in weight gain in mice from these different vendors during high fat diet stress. However, neither specific, highly divergent members of the gut microbiota (Lactobacillus murinus, segmented filamentous bacteria) nor the horizontally transmissible gut microbiota were found to be responsible. Constitutive differences in locomotor activity were observed, however. These data underscore the importance of selecting appropriate controls in this widely used model of human obesity.

3.
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
4.
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
5.
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
6.
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
7.
Adipocyte ; 1(4): 203-214, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23700535

ABSTRACT

PKCλ, an atypical member of the multifunctional protein kinase C family, has been implicated in the regulation of insulin-stimulated glucose transport and of the intracellular immune response. To further elucidate the role of this cellular regulator in diet-induced obesity and insulin resistance, we generated both liver (PKC-Alb) and adipose tissue (PKC-Ap2) specific knockout mice. Body weight, fat mass, food intake, glucose homeostasis and energy expenditure were evaluated in mice maintained on either chow or high fat diet (HFD). Ablation of PKCλ from the adipose tissue resulted in mice that were indistinguishable from their wild-type littermates. However, PKC-Alb mice were resistant to diet-induced obesity (DIO). Surprisingly this DIO resistance was not associated with either a reduction in caloric intake or an increase in energy expenditure as compared with their wild-type littermates. Furthermore, these mice displayed an improvement in glucose tolerance. When maintained on chow diet, these mice were similar to wild types in respect to body weight and fat mass, yet insulin sensitivity was impaired compared with wt littermates. Taken together these data suggest that hepatic PKCλ is modulating insulin-mediated glucose turnover and response to high fat diet feeding, thus offering a deeper understanding of an important target for anti-obesity therapeutics.

8.
Nat Chem Biol ; 5(10): 749-57, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19597507

ABSTRACT

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.


Subject(s)
Glucagon-Like Peptide 1/agonists , Obesity/drug therapy , Peptides, Cyclic/therapeutic use , Polyethylene Glycols/chemistry , Receptors, Glucagon/agonists , Adipose Tissue/drug effects , Amino Acid Sequence , Animals , Body Weight/drug effects , Cyclic AMP/biosynthesis , Eating/drug effects , Energy Metabolism/drug effects , Glucose Tolerance Test , Mice , Mice, Obese , Models, Molecular , Molecular Sequence Data , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Protein Conformation
SELECTION OF CITATIONS
SEARCH DETAIL
...