Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Mol Metab ; 55: 101392, 2022 01.
Article in English | MEDLINE | ID: mdl-34781035

ABSTRACT

OBJECTIVE: Obesity-linked type 2 diabetes (T2D) is a worldwide health concern and many novel approaches are being considered for its treatment and subsequent prevention of serious comorbidities. Co-administration of glucagon like peptide 1 (GLP-1) and peptide YY3-36 (PYY3-36) renders a synergistic decrease in energy intake in obese men. However, mechanistic details of the synergy between these peptide agonists and their effects on metabolic homeostasis remain relatively scarce. METHODS: In this study, we utilized long-acting analogues of GLP-1 and PYY3-36 (via Fc-peptide conjugation) to better characterize the synergistic pharmacological benefits of their co-administration on body weight and glycaemic regulation in obese and diabetic mouse models. Hyperinsulinemic-euglycemic clamps were used to measure weight-independent effects of Fc-PYY3-36 + Fc-GLP-1 on insulin action. Fluorescent light sheet microscopy analysis of whole brain was performed to assess activation of brain regions. RESULTS: Co-administration of long-acting Fc-IgG/peptide conjugates of Fc-GLP-1 and Fc-PYY3-36 (specific for PYY receptor-2 (Y2R)) resulted in profound weight loss, restored glucose homeostasis, and recovered endogenous ß-cell function in two mouse models of obese T2D. Hyperinsulinemic-euglycemic clamps in C57BLKS/J db/db and diet-induced obese Y2R-deficient (Y2RKO) mice indicated Y2R is required for a weight-independent improvement in peripheral insulin sensitivity and enhanced hepatic glycogenesis. Brain cFos staining demonstrated distinct temporal activation of regions of the hypothalamus and hindbrain following Fc-PYY3-36 + Fc-GLP-1R agonist administration. CONCLUSIONS: These results reveal a therapeutic approach for obesity/T2D that improved insulin sensitivity and restored endogenous ß-cell function. These data also highlight the potential association between the gut-brain axis in control of metabolic homeostasis.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Peptide YY/metabolism , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/metabolism , Diet , Eating/drug effects , Energy Intake/drug effects , Energy Metabolism/drug effects , Gastric Bypass , Glucagon-Like Peptide-1 Receptor/metabolism , Hypothalamus , Insulin Resistance/physiology , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/physiopathology , Peptide YY/physiology , Weight Loss
2.
World J Gastroenterol ; 25(33): 4904-4920, 2019 Sep 07.
Article in English | MEDLINE | ID: mdl-31543682

ABSTRACT

BACKGROUND: The trans-fat containing AMLN (amylin liver non-alcoholic steatohepatitis, NASH) diet has been extensively validated in C57BL/6J mice with or without the Lepob/Lepob (ob/ob) mutation in the leptin gene for reliably inducing metabolic and liver histopathological changes recapitulating hallmarks of NASH. Due to a recent ban on trans-fats as food additive, there is a marked need for developing a new diet capable of promoting a compatible level of disease in ob/ob and C57BL/6J mice. AIM: To develop a biopsy-confirmed mouse model of NASH based on an obesogenic diet with trans-fat substituted by saturated fat. METHODS: Male ob/ob mice were fed AMLN diet or a modified AMLN diet with trans-fat (Primex shortening) substituted by equivalent amounts of palm oil [Gubra amylin NASH, (GAN) diet] for 8, 12 and 16 wk. C57BL/6J mice were fed the same diets for 28 wk. AMLN and GAN diets had similar caloric content (40% fat kcal), fructose (22%) and cholesterol (2%) level. RESULTS: The GAN diet was more obesogenic compared to the AMLN diet and impaired glucose tolerance. Biopsy-confirmed steatosis, lobular inflammation, hepatocyte ballooning, fibrotic liver lesions and hepatic transcriptome changes were similar in ob/ob mice fed the GAN or AMLN diet. C57BL/6J mice developed a mild to moderate fibrotic NASH phenotype when fed the same diets. CONCLUSION: Substitution of Primex with palm oil promotes a similar phenotype of biopsy-confirmed NASH in ob/ob and C57BL/6J mice, making GAN diet-induced obese mouse models suitable for characterizing novel NASH treatments.


Subject(s)
Disease Models, Animal , Liver/pathology , Non-alcoholic Fatty Liver Disease/etiology , Palm Oil/adverse effects , Animals , Biopsy , Diet, High-Fat/adverse effects , Humans , Leptin/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/pathology , Trans Fatty Acids/adverse effects
3.
Nat Neurosci ; 13(7): 877-82, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20526334

ABSTRACT

Cholesterol circulates in the blood in association with triglycerides and other lipids, and elevated blood low-density lipoprotein cholesterol carries a risk for metabolic and cardiovascular disorders, whereas high-density lipoprotein (HDL) cholesterol in the blood is thought to be beneficial. Circulating cholesterol is the balance among dietary cholesterol absorption, hepatic synthesis and secretion, and the metabolism of lipoproteins by various tissues. We found that the CNS is also an important regulator of cholesterol in rodents. Inhibiting the brain's melanocortin system by pharmacological, genetic or endocrine mechanisms increased circulating HDL cholesterol by reducing its uptake by the liver independent of food intake or body weight. Our data suggest that a neural circuit in the brain is directly involved in the control of cholesterol metabolism by the liver.


Subject(s)
Cholesterol, HDL/blood , Ghrelin/physiology , Hypothalamus/metabolism , Liver/metabolism , Melanocortins/metabolism , Animals , Body Weight , CD36 Antigens/metabolism , Eating , Ghrelin/genetics , Glucagon-Like Peptide 1/physiology , Homeostasis/physiology , Mice , Mice, Knockout , Neurosecretory Systems/metabolism , Rats , Rats, Wistar , Receptors, Melanocortin/genetics , Receptors, Melanocortin/physiology , Scavenger Receptors, Class B/metabolism
4.
Trends Endocrinol Metab ; 21(8): 473-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20413324

ABSTRACT

Although the adipokine leptin is regarded as the prototypical long-term signal of energy balance, obese individuals are largely nonresponsive to exogenous leptin administration. Restoration of leptin responsiveness in obesity has been elusive despite a detailed understanding of the molecular mechanisms of leptin signaling. Recent translational research findings point to a potential therapeutic approach that incorporates amylin (a beta-cell hormone) and leptin agonism, with amylin restoring or enhancing leptin sensitivity. Here we hypothesize various physiological, neurobiological and molecular mechanisms that could mediate the interaction of these two neurohormonal signals and discuss several methodological challenges. Understanding how amylin agonism improves leptin function could point to general therapeutic strategies for combating leptin resistance and associated obesity.


Subject(s)
Islet Amyloid Polypeptide/pharmacology , Islet Amyloid Polypeptide/physiology , Leptin/pharmacology , Leptin/physiology , Obesity/drug therapy , Animals , Body Weight/drug effects , Drug Synergism , Humans , Hypothalamus/drug effects , Islet Amyloid Polypeptide/therapeutic use , Leptin/therapeutic use , Obesity/physiopathology , Rhombencephalon/drug effects
5.
Endocrinology ; 149(11): 5679-87, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18669592

ABSTRACT

Previously, we reported that combination treatment with rat amylin (100 microg/kg.d) and murine leptin (500 microg/kg.d) elicited greater inhibition of food intake and greater body weight loss in diet-induced obese rats than predicted by the sum of the monotherapy conditions, a finding consistent with amylin-induced restoration of leptin responsiveness. In the present study, a 3 x 4 factorial design was used to formally test for a synergistic interaction, using lower dose ranges of amylin (0, 10, and 50 microg/kg.d) and leptin (0, 5, 25, and 125 microg/kg.d), on food intake and body weight after 4 wk continuous infusion. Response surface methodology analysis revealed significant synergistic anorexigenic (P < 0.05) and body weight-lowering (P < 0.05) effects of amylin/leptin combination treatment, with up to 15% weight loss at doses considerably lower than previously reported. Pair-feeding (PF) experiments demonstrated that reduction of food intake was the predominant mechanism for amylin/leptin-mediated weight loss. However, fat loss was 2-fold greater in amylin/leptin-treated rats than PF controls. Furthermore, amylin/leptin-mediated weight loss was not accompanied by the counterregulatory decrease in energy expenditure and chronic shift toward carbohydrate (rather than fat) utilization observed with PF. Hepatic gene expression analyses revealed that 28 d treatment with amylin/leptin (but not PF) was associated with reduced expression of genes involved in hepatic lipogenesis (Scd1 and Fasn mRNA) and increased expression of genes involved in lipid utilization (Pck1 mRNA). We conclude that amylin/leptin interact synergistically to reduce body weight and adiposity in diet-induced obese rodents through a number of anorexigenic and metabolic effects.


Subject(s)
Amyloid/pharmacology , Drug Resistance/drug effects , Leptin/pharmacology , Obesity/pathology , Signal Transduction/drug effects , Amyloid/administration & dosage , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Body Weight/drug effects , Caloric Restriction , Diet/adverse effects , Drug Evaluation, Preclinical , Drug Synergism , Eating/drug effects , Islet Amyloid Polypeptide , Leptin/administration & dosage , Lipids/blood , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Rats , Rats, Sprague-Dawley
6.
Proc Natl Acad Sci U S A ; 105(20): 7257-62, 2008 May 20.
Article in English | MEDLINE | ID: mdl-18458326

ABSTRACT

Body weight is regulated by complex neurohormonal interactions between endocrine signals of long-term adiposity (e.g., leptin, a hypothalamic signal) and short-term satiety (e.g., amylin, a hindbrain signal). We report that concurrent peripheral administration of amylin and leptin elicits synergistic, fat-specific weight loss in leptin-resistant, diet-induced obese rats. Weight loss synergy was specific to amylin treatment, compared with other anorexigenic peptides, and dissociable from amylin's effect on food intake. The addition of leptin after amylin pretreatment elicited further weight loss, compared with either monotherapy condition. In a 24-week randomized, double-blind, clinical proof-of-concept study in overweight/obese subjects, coadministration of recombinant human leptin and the amylin analog pramlintide elicited 12.7% mean weight loss, significantly more than was observed with either treatment alone (P < 0.01). In obese rats, amylin pretreatment partially restored hypothalamic leptin signaling (pSTAT3 immunoreactivity) within the ventromedial, but not the arcuate nucleus and up-regulated basal and leptin-stimulated signaling in the hindbrain area postrema. These findings provide both nonclinical and clinical evidence that amylin agonism restored leptin responsiveness in diet-induced obesity, suggesting that integrated neurohormonal approaches to obesity pharmacotherapy may facilitate greater weight loss by harnessing naturally occurring synergies.


Subject(s)
Amyloid/agonists , Amyloid/chemistry , Leptin/metabolism , Adipose Tissue/metabolism , Amyloid/metabolism , Amyloid/pharmacology , Animals , Body Weight , Caloric Restriction , Disease Models, Animal , Hormones/metabolism , Hypothalamus/metabolism , Islet Amyloid Polypeptide , Leptin/analogs & derivatives , Leptin/pharmacology , Models, Biological , Obesity/genetics , Obesity/therapy , Oxygen Consumption , Rats
SELECTION OF CITATIONS
SEARCH DETAIL