Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
Am J Physiol Endocrinol Metab ; 303(2): E265-71, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-22621866

ABSTRACT

Glucagon-like peptide-1 (GLP-1) and oxyntomodulin (OXM) are peptide hormones secreted postprandially from the gut that stimulate insulin secretion in a glucose-dependent manner. OXM activates both the GLP-1 receptor (GLP1R) and the glucagon receptor (GCGR). It has been suggested that OXM acutely modulates glucose metabolism solely through GLP1R agonism. Because OXM activates the GLP1R with lower affinity than GLP-1, we generated a peptide analog (Q→E, OXMQ3E) that does not exhibit glucagon receptor agonist activity but retains the same affinity as OXM for GLP1R. We compared the effects of OXM and OXMQ3E in a glucose tolerance test and, to better characterize the effect on glucose metabolism, we performed controlled infusions of OXM or OXMQ3E during a hyperglycemic clamp performed in wild-type, Glp1r(-/-), and Gcgr(-/-) mice. Our findings show that OXM, but not OXMQ3E, activates the GCGR in vivo. Second, OXM and OXMQ3E improve glucose tolerance following an acute glucose challenge and during a hyperglycemic clamp in mice. Finally, OXM infusion during a glucose clamp reduces the glucose infusion rate (GIR) despite a simultaneous increase in insulin levels in Glp1r(-/-) mice, whereas OXM and OXMQ3E increase GIR to a similar extent in Gcgr(-/-) mice. In conclusion, activation of the GCGR seems to partially attenuate the acute beneficial effects on glucose and contributes to the insulinotropic action of oxyntomodulin.


Subject(s)
Glucagon-Like Peptide 1/pharmacology , Glucose/metabolism , Oxyntomodulin/pharmacology , Animals , Blood Glucose/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose Clamp Technique , Glucose Tolerance Test , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism
2.
Obesity (Silver Spring) ; 20(8): 1566-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22421924

ABSTRACT

Oxyntomodulin (OXM) is a peptide secreted postprandially from the L-cells of the gut that has a weak affinity for both the glucagon-like peptide-1 receptor (GLP1R) and the glucagon receptor (GCGR). Peripheral administration of OXM in humans and rodents causes weight loss reducing food intake and increasing energy expenditure. It has been suggested that OXM modulates energy intake solely through GLP1R agonism. Because glucagon decreases food intake in rodents and humans, we examined whether activation of the GCGR is involved in the body weight-lowering effects of OXM. We identified an equipotent GLP1R-selective peptide agonist that differs from OXM by only one residue (Q3→E, OXMQ3E), but has no significant GCGR agonist activity in vitro and ~100-fold reduced ability to stimulate liver glycogenolysis. Chronic treatment of obese mice with OXM and OXMQ3E demonstrated that OXM exhibits superior weight loss and lipid-lowering efficacy, and antihyperglycemic activity that is comparable to the corresponding GLP1R-selective agonist. Studies in Glp1r(-/-) mice and coadministration of OXM and a GCGR antagonist revealed that the antiobesity effect of OXM requires activation of both GLP1R and GCGR. Our data provide new insight into the mechanism of action of OXM and suggest that activation of GCGR is involved in the body weight-lowering action of OXM.


Subject(s)
Anti-Obesity Agents/pharmacology , Energy Intake/physiology , Glucagon/metabolism , Obesity/metabolism , Oxyntomodulin/metabolism , Receptors, Glucagon/agonists , Weight Loss/drug effects , Animals , Energy Intake/drug effects , Glucagon-Like Peptide-1 Receptor , Glycogenolysis/drug effects , Humans , Hypoglycemic Agents/pharmacology , Hypolipidemic Agents/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/drug therapy , Oxyntomodulin/pharmacology , Receptors, Glucagon/metabolism
3.
PLoS One ; 7(4): e35240, 2012.
Article in English | MEDLINE | ID: mdl-22506074

ABSTRACT

Short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate, are metabolites formed by gut microbiota from complex dietary carbohydrates. Butyrate and acetate were reported to protect against diet-induced obesity without causing hypophagia, while propionate was shown to reduce food intake. However, the underlying mechanisms for these effects are unclear. It was suggested that SCFAs may regulate gut hormones via their endogenous receptors Free fatty acid receptors 2 (FFAR2) and 3 (FFAR3), but direct evidence is lacking. We examined the effects of SCFA administration in mice, and show that butyrate, propionate, and acetate all protected against diet-induced obesity and insulin resistance. Butyrate and propionate, but not acetate, induce gut hormones and reduce food intake. As FFAR3 is the common receptor activated by butyrate and propionate, we examined these effects in FFAR3-deficient mice. The effects of butyrate and propionate on body weight and food intake are independent of FFAR3. In addition, FFAR3 plays a minor role in butyrate stimulation of Glucagon-like peptide-1, and is not required for butyrate- and propionate-dependent induction of Glucose-dependent insulinotropic peptide. Finally, FFAR3-deficient mice show normal body weight and glucose homeostasis. Stimulation of gut hormones and food intake inhibition by butyrate and propionate may represent a novel mechanism by which gut microbiota regulates host metabolism. These effects are largely intact in FFAR3-deficient mice, indicating additional mediators are required for these beneficial effects.


Subject(s)
Butyrates/metabolism , Fatty Acids, Nonesterified/metabolism , Gastrointestinal Hormones/metabolism , Obesity/metabolism , Propionates/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Body Weight/drug effects , Body Weight/physiology , Butyrates/pharmacology , Diet , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Homeostasis/drug effects , Homeostasis/physiology , Insulin Resistance/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/prevention & control , Propionates/pharmacology
4.
Endocrinology ; 152(7): 2644-54, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21586559

ABSTRACT

Neuromedin U (NMU) is a highly conserved peptide reported to modulate energy homeostasis. Pharmacological studies have shown that centrally administered NMU inhibits food intake, reduces body weight, and increases energy expenditure. NMU-deficient mice develop obesity, whereas transgenic mice overexpressing NMU become lean and hypophagic. Two high-affinity NMU receptors, NMUR1 and NMUR2, have been identified. NMUR1 is found primarily in the periphery and NMUR2 primarily in the brain, where it mediates the anorectic effects of centrally administered NMU. Given the broad expression pattern of NMU, we evaluated whether peripheral administration of NMU has effects on energy homeostasis. We observed that acute and chronic peripheral administration of NMU in rodents dose-dependently reduced food intake and body weight and that these effects required NMUR1. The anorectic effects of NMU appeared to be partly mediated by vagal afferents. NMU treatment also increased core body temperature and metabolic rate in mice, suggesting that peripheral NMU modulates energy expenditure. Additionally, peripheral administration of NMU significantly improved glucose excursion. Collectively, these data suggest that NMU functions as a peripheral regulator of energy and glucose homeostasis and the development of NMUR1 agonists may be an effective treatment for diabetes and obesity.


Subject(s)
Energy Metabolism , Glucose Intolerance/prevention & control , Neuropeptides/physiology , Animals , Appetite Regulation , Basal Metabolism , Body Temperature Regulation , Diabetes Mellitus/drug therapy , Food Preferences , Isoenzymes/genetics , Isoenzymes/metabolism , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Molecular Targeted Therapy , Neuropeptides/administration & dosage , Obesity/drug therapy , Rats , Rats, Sprague-Dawley , Receptors, Neurotransmitter/genetics , Receptors, Neurotransmitter/metabolism , Weight Loss
SELECTION OF CITATIONS
SEARCH DETAIL