Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 3(11): 100810, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36384093

ABSTRACT

Glucagon analogs show promise as components of next-generation, multi-target, anti-obesity therapeutics. The biology of chronic glucagon treatment, in particular, its ability to induce energy expenditure and weight loss, remains poorly understood. Using a long-acting glucagon analog, G108, we demonstrate that glucagon-mediated body weight loss is intrinsically linked to the hypoaminoacidemia associated with its known amino acid catabolic action. Mechanistic studies reveal an energy-consuming response to low plasma amino acids in G108-treated mice, prevented by dietary amino acid supplementation and mimicked by a rationally designed low amino acid diet. Therefore, low plasma amino acids are a pre-requisite for G108-mediated energy expenditure and weight loss. However, preventing hypoaminoacidemia with additional dietary protein does not affect the ability of G108 to improve glycemia or hepatic steatosis in obese mice. These studies provide a mechanism for glucagon-mediated weight loss and confirm the hepatic glucagon receptor as an attractive molecular target for metabolic disease therapeutics.


Subject(s)
Glucagon , Weight Loss , Mice , Animals , Glucagon/metabolism , Energy Metabolism/physiology , Receptors, Glucagon/metabolism , Mice, Obese , Amino Acids/pharmacology
2.
Sci Rep ; 11(1): 22577, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799628

ABSTRACT

Glucagon receptor agonists show promise as components of next generation metabolic syndrome pharmacotherapies. However, the biology of glucagon action is complex, controversial, and likely context dependent. As such, a better understanding of chronic glucagon receptor (GCGR) agonism is essential to identify and mitigate potential clinical side-effects. Herein we present a novel, long-acting glucagon analogue (GCG104) with high receptor-specificity and potent in vivo action. It has allowed us to make two important observations about the biology of sustained GCGR agonism. First, it causes weight loss in mice by direct receptor signalling at the level of the liver. Second, subtle changes in GCG104-sensitivity, possibly due to interindividual variation, may be sufficient to alter its effects on metabolic parameters. Together, these findings confirm the liver as a principal target for glucagon-mediated weight loss and provide new insights into the biology of glucagon analogues.


Subject(s)
Anti-Obesity Agents/pharmacology , Glucagon/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Receptors, Glucagon/agonists , Weight Loss/drug effects , Animals , Anti-Obesity Agents/pharmacokinetics , Biological Variation, Population , Dose-Response Relationship, Drug , Female , Glucagon/analogs & derivatives , Glucagon/pharmacokinetics , HEK293 Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Rats, Wistar , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Signal Transduction
3.
J Cachexia Sarcopenia Muscle ; 11(5): 1187-1199, 2020 10.
Article in English | MEDLINE | ID: mdl-32274897

ABSTRACT

BACKGROUND: Constitutional thinness (CT), a non-malnourished underweight state with no eating disorders, is characterized by weight gain resistance to high fat diet. Data issued from muscle biopsies suggested blunted anabolic mechanisms in free-living state. Weight and metabolic responses to protein caloric supplementation has not been yet explored in CT. METHODS: A 2 week overfeeding (additional 600 kcal, 30 g protein, 72 g carbohydrate, and 21 g fat) was performed to compare two groups of CTs (12 women and 11 men) to normal-weight controls (12 women and 10 men). Bodyweight, food intake, energy expenditure, body composition, nitrogen balance, appetite hormones profiles, and urine metabolome were monitored before and after overfeeding. RESULTS: Before overfeeding, positive energy gap was found in both CT genders (309 ± 370 kcal in CT-F and 332 ± 709 kcal in CT-M) associated with higher relative protein intake per kilo (1.74 ± 0.32 g/kg/day in CT-F vs. 1.16 ± 0.23 in C-F, P < 0.0001; 1.56 ± 0.36 in CT-M vs. 1.22 ± 0.32 in C-M, P = 0.03), lower nitrogen (7.26 ± 2.36 g/day in CT-F vs. 11.41 ± 3.64 in C-F, P = 0.003; 9.70 ± 3.85 in CT-M vs. 14.14 ± 4.19 in C-M, P = 0.02), but higher essential amino acids urinary excretion (CT/C fold change of 1.13 for leucine and 1.14 for arginine) in free-living conditions. After overfeeding, CTs presented an accentuated positive energy gap, still higher than in controls (675 ± 540 in CTs vs. 379 ± 427 in C, P = 0.04). Increase in lean mass was induced in both controls genders but not in CTs (a trend was noticed in CT women), despite a similar nitrogen balance after overfeeding (5.06 ± 4.33 g/day in CTs vs. 4.28 ± 3.15 in controls, P = 0.49). Higher anorectic gut hormones' tone, glucagon-like peptide 1 and peptide tyrosine tyrosine, during test meal and higher snacking frequency were noticed before and after overfeeding in CTs. CONCLUSIONS: The blunted muscle energy mechanism, previously described in CTs in free-living state, is associated with basal saturated protein turn over suggested by the concordance of positive nitrogen balance and an increased urine excretion of several essential amino acids. This saturation cannot be overpassed by increasing this spontaneous high-protein intake suggesting a resistance to lean mass gain in CT phenotype.


Subject(s)
Social Conditions , Thinness , Adolescent , Body Composition , Energy Metabolism , Female , Humans , Male , Weight Gain , Young Adult
4.
Am J Physiol Gastrointest Liver Physiol ; 316(3): G366-G371, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30576216

ABSTRACT

Activation of ghrelin is controlled by the enzyme ghrelin- O-acyl transferase (GOAT). In humans, localization of this acylation is poorly understood. The aim of this study is to explore GOAT localization and activation in the human liver by evaluating both bioactive and non-bioactive ghrelin in the bloodstream entering and leaving the liver and to simultaneously evaluate GOAT mRNA expression in the liver. A healthy part of oncologic hepatic tissue collected from nine patients undergoing hepatectomy was used to evaluate GOAT mRNA expression by quantitative real-time polymerase chain reaction (RT-qPCR). Simultaneously, blood from the portal vein, the suprahepatic vein, the subclavicular vein, and the radial artery was also sampled to assay total and acylated ghrelin. Acylated ghrelin level was significantly increased in the suprahepatic vein compared with the portal vein level (385 ± 42 ng/ml vs. 268 ± 24 ng/ml, P = 0.04). Suprahepatic-to-portal vein ratio for acylated ghrelin (acylation ratio) is 1.4 ± 0.1. Mean expression of GOAT mRNA in the liver, expressed as 2-∆Ct·µg total RNA-1·1 µl of liver tissue-1 was at 0.042 ± 0.021 arbitrary units. GOAT mRNA expression in the liver was correlated with acylated-to-total ghrelin ratio in the suprahepatic vein ( P = 0.016, R = 0.75) and with the acylation liver ratio ( P = 0.05, R = 0.61). Blood concentration of acylated ghrelin was found significantly increased after its passage through the liver, suggesting that acylation can occur in the liver. RT-qPCR data confirmed the presence of GOAT in the liver, with a positive correlation between GOAT expression and acylated ghrelin liver ratio. This study strongly suggests that the liver is a site of ghrelin acylation in humans. NEW & NOTEWORTHY Although the activation of ghrelin by the enzyme ghrelin- O-acyl transferase (GOAT) is yet well demonstrated, its localization, especially in humans, remains poorly understood. We explored GOAT localization and activation in the human liver by simultaneously evaluating both bioactive and non-bioactive ghrelin in the bloodstream entering and leaving the liver and also GOAT mRNA expression in the liver. We therefore showed for the first time, to our knowledge, that GOAT localized in the liver is active and takes part in ghrelin activation.


Subject(s)
Acylation/physiology , Acyltransferases/metabolism , Ghrelin/metabolism , Liver/metabolism , Acyltransferases/genetics , Adult , Female , Gastric Mucosa/metabolism , Gene Expression Regulation, Enzymologic/genetics , Humans , Liver Neoplasms/metabolism , Male , Middle Aged , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction/methods
5.
Psychoneuroendocrinology ; 84: 94-100, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28692876

ABSTRACT

INTRODUCTION: Constitutional thinness (CT) is an underweight state characterized by normal menstruations and no change in feeding behaviour. Thinness is the only resemblance between Anorexia Nervosa (AN) and CT. Removal of amenorrhea from the new DSM 5 definition of AN might result in misdiagnosis between these two populations. The objective of this study was to compare CT, AN and Control subjects in terms of biological, anthropometric, and psychological markers in order to better distinguish AN from CT subjects. MATERIALS AND METHODS: Body composition, nutritional markers, pituitary hormones, bone markers and psychological scores were evaluated in three groups of young women: fifty-six CT, forty restrictive-type AN and fifty-four Control subjects. For every marker, a receiver Operator Characteristics (ROC) curve was calculated to evaluate the accuracy of differentiation between AN and CT groups. RESULTS: For most studied parameters, CT subjects were similar to Controls but dramatically different from AN subjects. DEBQ Restrained Eating subscale score was identified by ROC data analysis as the only psychological parameter tested to successfully differentiate AN from CT. Free-T3 and Leptin were shown to be powerful markers to differentiate AN and CT populations as they were highly specific and sensitive ones. CONCLUSION: The exclusive use of psychological testing criteria is not always sufficient to differentiate AN and CT patients. Minimally, additional testing of Free T3 levels, which is cheap and widely accessible for general practitioners, should be completed to avoid misdiagnosis which could result in the implementation of ineffective treatment plans and social stigmatization for CT women.


Subject(s)
Anorexia Nervosa/diagnosis , Anorexia Nervosa/psychology , Thinness/diagnosis , Adult , Anthropometry/methods , Biomarkers , Body Composition , Body Mass Index , Body Weight , Diagnostic and Statistical Manual of Mental Disorders , Female , Humans , Leptin/analysis , Leptin/blood , Thyroid Hormones/analysis , Thyroid Hormones/blood
6.
Pancreas ; 45(7): 967-73, 2016 08.
Article in English | MEDLINE | ID: mdl-26731187

ABSTRACT

OBJECTIVES: Reports have suggested a link between treatment with glucagon-like peptide 1 (GLP-1) analogs and an increased risk of pancreatitis. Oxyntomodulin, a dual agonist of both GLP-1 and glucagon receptors, is currently being investigated as a potential antiobesity therapy, but little is known about its pancreatic safety. The aim of the study was to investigate the acute effect of oxyntomodulin and other proglucagon-derived peptides on the rat exocrine pancreas. METHODS: Glucagon-like peptide 1, oxyntomodulin, glucagon, and exendin-4 were infused into anesthetized rats to measure plasma amylase concentration changes. In addition, the effect of each peptide on both amylase release and proliferation in rat pancreatic acinar (AR42J) and primary isolated ductal cells was determined. RESULTS: Plasma amylase did not increase postpeptide infusion, compared with vehicle and cholecystokinin; however, oxyntomodulin inhibited plasma amylase when coadministered with cholecystokinin. None of the peptides caused a significant increase in proliferation rate or amylase secretion from acinar and ductal cells. CONCLUSIONS: The investigated peptides do not have an acute effect on the exocrine pancreas with regard to proliferation and plasma amylase, when administered individually. Oxyntomodulin seems to be a potent inhibitor of amylase release, potentially making it a safer antiobesity agent regarding pancreatitis, compared with GLP-1 agonists.


Subject(s)
Oxyntomodulin/pharmacology , Pancreas, Exocrine/drug effects , Peptides/pharmacology , Proglucagon/pharmacology , Actins/genetics , Amylases/blood , Amylases/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Exenatide , Gene Expression , Glucagon/administration & dosage , Glucagon/pharmacology , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/pharmacology , Injections, Intravenous , Ki-67 Antigen/genetics , Male , Oxyntomodulin/administration & dosage , Pancreas, Exocrine/metabolism , Peptides/administration & dosage , Proglucagon/administration & dosage , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Venoms/administration & dosage , Venoms/pharmacology
7.
Peptides ; 73: 95-100, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26431789

ABSTRACT

Oxyntomodulin analogues offer a novel treatment for obesity. However during analogue screening in a rat model increased food intake was consistently observed. To further investigate this finding, a series of representative analogues (OXM14 and OXM15) and their Glu-3 equivalents (OXM14E3 and OXM15E3) were administered to rats for 7 days and food intake and bodyweight measurements taken. To investigate the role of glucagon receptor activation glutamate (Glu/E) was substituted at amino acid position 3. GLP-1 and glucagon receptor efficacy of the oxyntomodulin analogues and their Glu-3 counterparts were measured at the rat receptors in vitro. Doses of 25 nmol/kg of OXM14 and OXM15 increased food intake by up to 20%. Bodyweight was not significantly increased. Food intake was not increased with the Glu-3 peptides, indicating that a glucagon receptor mechanism may be responsible for the increase in food intake.


Subject(s)
Body Weight/drug effects , Eating/drug effects , Oxyntomodulin/pharmacology , Animals , CHO Cells , Cricetinae , Cricetulus , Male , Oxyntomodulin/chemistry , Rats , Rats, Wistar
8.
Curr Ther Res Clin Exp ; 77: 111-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26843896

ABSTRACT

AIMS: To investigate the effect of Glu-3 OXM-like analogues on food intake and bodyweight in male rats. BACKGROUND: Oxyntomodulin (OXM) is a natural agonist at both the glucagon receptor (GCGr) and the glucagon-like peptide 1 receptor (GLP-1r), and peripheral administration reduces food intake and increases energy expenditure in rodents and humans. Substituting the native glutamine (Gln) at amino acid position 3 of OXM for glutamate (Glu) has previously been shown to diminish GCGr activity without affecting GLP-1r activity. The effects of Glu-3 OXM analogues have not been investigated in rats. METHODS: The effect of 2 Glu-3-substituted OXM-like analogues (eg, OXM14E3 and OXM15E3) on food intake and body weight was investigated in male Wistar rats during 6 days of daily subcutaneous (SC) administration. The effects of Glu-3 substitution on analogue binding and activity at the rat GCGr and rat GLP-1 receptor were investigated in vitro using Chinese hamster ovary or Chinese hamster lung cells. RESULTS: We report the novel finding that 2 5-nmol/kg Glu-3 OXM-like analogues (OXM14E3 and OXM15E3) significantly increased rat body weight by up to 4% compared with the equivalent non-Glu-3 analogues (OXM14 and OXM15), without affecting food intake. The effect of OXM15E3 on body weight was dose-dependent. Glu-3 analogues, including Glu-3 OXM, decreased glucagon-mediated cyclic adenosine monophosphate accumulation in Chinese hamster ovary cells expressing the rat GCGr, suggesting they may be acting as antagonists. CONCLUSIONS: The results indicate Glu-3 OXM-like analogues might not be suitable tools to investigate the mechanism of OXM analogue action in a rat model because they significantly increase body weight independent of food intake. Glu-3 OXM analogues are partial agonists at the rat GCGr and may also act as antagonists, possibly resulting in the observed increase in body weight.

9.
Peptides ; 39: 6-10, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23123314

ABSTRACT

Pancreatic polypeptide (PP), peptide YY (PYY) and neuropeptide Y (NPY), members of the PP-fold family share a high degree of sequence homology. Nuclear magnetic resonance (NMR) and X-ray crystallography studies have shown these peptides can adopt a tightly organized tertiary structure called the PP-fold, which has long been assumed to be the active structure of this family of peptides. To date, however, no studies have been completed with PYY and PP which confirm if the PP-fold structure is important for their physiological actions. The aim of the study was to test if PYY and PP locked into the PP-fold maintained biological activity. Therefore, we designed and produced analogs of PP and PYY in a cyclic conformation with two cysteine amino acid substitutions at the N-terminus and at position 27. These were oxidized to form a cysteine disulfide bond locking the peptides into the PP-fold structure. Studies demonstrate that the cyclic analogs have both similar in vivo activity to their parent molecules, and affinity for the Y2 and Y4 receptors. Results suggest that the proposed PP and PYY-fold is likely to be their biologically active conformation.


Subject(s)
Cystine/chemistry , Eating/drug effects , Pancreatic Polypeptide/chemistry , Peptide YY/chemistry , Peptides, Cyclic/chemistry , Animals , Cystine/chemical synthesis , Food Deprivation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Pancreatic Polypeptide/pharmacology , Peptide YY/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Receptors, Neuropeptide Y/chemistry
10.
Br J Clin Pharmacol ; 73(2): 232-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21834938

ABSTRACT

AIMS: The objectives of this phase 1 study were to confirm the tolerability of single ascending subcutaneous doses of PP 1420 in healthy subjects, to assess its adverse effects and to investigate the drug's pharmacokinetics and dose proportionality. METHODS: This was a double-blind, placebo-controlled, randomized study. There were three dosing periods. Each subject (n= 12) was randomized to receive one dose of placebo and two ascending doses of PP 1420, given as a subcutaneous injection. Blood samples were taken over 24 h to assess pharmacokinetics. Standard safety and laboratory data were collected. The primary endpoint was the tolerability of PP 1420. The secondary endpoint was exposure to PP 1420 as assessed by C(max) and AUC(0,∞). RESULTS: PP 1420 was well tolerated by all subjects with no serious adverse effects. Following single subcutaneous doses of PP 1420 at 2, 4 and 8 mg to male subjects, C(max) was reached at a median t(max) of approximately 1 h post dose (range 0.32-2.00 h). Thereafter, plasma concentrations of PP 1420 declined with geometric mean apparent terminal elimination t(1/2) ranging from 2.42-2.61 h (range 1.64-3.95 h) across all dose levels. CONCLUSIONS: Subcutaneous PP 1420 was well tolerated in healthy human subjects at single doses between 2-8 mg, with no tolerability issues arising. Where observed, adverse events were not serious, and there was no evidence of a dose-relationship to frequency of adverse events. The results therefore support the conduct of clinical trials to investigate efficacy, tolerability and pharmacokinetics during repeated dosing.


Subject(s)
Appetite Depressants/pharmacokinetics , Appetite/drug effects , Obesity/prevention & control , Pancreatic Polypeptide/analogs & derivatives , Receptors, Neuropeptide Y/agonists , Adolescent , Adult , Appetite Depressants/adverse effects , Area Under Curve , Dose-Response Relationship, Drug , Double-Blind Method , Humans , Injections, Subcutaneous , Male , Middle Aged , Pancreatic Polypeptide/adverse effects , Pancreatic Polypeptide/pharmacokinetics , United Kingdom , Young Adult
11.
Endocrinology ; 152(12): 4630-40, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21952244

ABSTRACT

Peptide YY(3-36) (PYY(3-36)) is a gut hormone that acts on Y2 receptors to reduce appetite. Obese humans are sensitive to the anorectic effects of PYY(3-36) and display a blunted postprandial rise in PYY(3-36). Bariatric surgery results in increased circulating PYY-immunoreactivity, which appears to play a role in postoperative weight loss. The utility of PYY(3-36) as an antiobesity treatment is limited by its short circulating half-life. Insight into the mechanisms by which PYY(3-36) is degraded may aid design of long-acting PYY(3-36) analogues or enzyme inhibitor therapies. We aimed to investigate the role of metalloendopeptidases in PYY(3-36) degradation and determine whether modulation of these enzymes enhanced PYY(3-36) plasma levels and bioactivity in vivo. Degradation and resultant cleavage products of PYY(3-36) were characterized after incubation with neprilysin and meprin ß and with a kidney brush border preparation in vitro. Specific metalloendopeptidase inhibitors were coadministered with PYY(3-36) to mice and subsequent PYY(3-36) plasma levels and bioactivity determined. Meprin ß cleaves PYY(3-36) at multiple conserved acidic sites. Blocking the actions of meprin ß prevents the degradative effect of kidney brush borders on PYY(3-36). In mice, pretreatment with actinonin significantly prolonged the anorectic effect of PYY(3-36) and maintained higher PYY(3-36) plasma levels than treatment with PYY(3-36) alone. These studies suggest that inhibiting the degradation of PYY(3-36) using specific inhibitor therapies and/or the design of analogues resistant to cleavage by meprins may be useful to antiobesity therapeutics.


Subject(s)
Metalloendopeptidases/metabolism , Peptide YY/metabolism , Animals , Anti-Obesity Agents , Enzyme Inhibitors/pharmacology , Gastrointestinal Hormones/metabolism , Half-Life , Kidney/metabolism , Mice , Peptide YY/antagonists & inhibitors , Peptide YY/blood , Peptide YY/chemistry , Protein Stability/drug effects , Tiopronin/pharmacology
12.
Endocr J ; 57(5): 359-72, 2010.
Article in English | MEDLINE | ID: mdl-20424341

ABSTRACT

The World Health Organisation has estimated that by 2015 approximately 2.3 billion adults will be overweight and more than 700 million obese. Obesity is associated with an increased risk of diabetes, cardiovascular events, stroke and cancer. The hypothalamus is a crucial region for integrating signals from central and peripheral pathways and plays a major role in appetite regulation. In addition, there are reciprocal connections with the brainstem and higher cortical centres. In the arcuate nucleus of the hypothalamus, there are two major neuronal populations which stimulate or inhibit food intake and influence energy homeostasis. Within the brainstem, the dorsal vagal complex plays a role in the interpretation and relaying of peripheral signals. Gut hormones act peripherally to modulate digestion and absorption of nutrients. However, they also act as neurotransmitters within the central nervous system to control food intake. Peptide YY, pancreatic polypeptide, glucagon-like peptide-1 and oxyntomodulin suppress appetite, whilst ghrelin increases appetite through afferent vagal fibres to the caudal brainstem or directly to the hypothalamus. A better understanding of the role of these gut hormones may offer the opportunity to develop successful treatments for obesity. Here we review the current understanding of the role of gut hormones and the hypothalamus on food intake and body weight control.


Subject(s)
Appetite Regulation/physiology , Gastrointestinal Hormones/physiology , Hypothalamus/physiology , Adult , Animals , Appetite Regulation/genetics , Brain Stem/metabolism , Brain Stem/physiology , Feeding Behavior/physiology , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiology , Humans , Models, Biological , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Reward
13.
Endocrinology ; 150(4): 1712-22, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19074579

ABSTRACT

Oxyntomodulin (Oxm) is an intestinal peptide that inhibits food intake and body weight in rodents and humans. These studies used peptide analogs to study aspects of structure and function of Oxm, and the sensitivity of parts of the Oxm sequence to degradation. Analogs of Oxm were synthesized and studied using receptor binding and degradation studies in vitro. Their effects on food intake and conditioned taste avoidance were measured in vivo in rodents. Oxm breakdown by the enzyme dipeptidyl peptidase IV (DPPIV) was demonstrated in vitro and in vivo. In vitro degradation was reduced and in vivo bioactivity increased by inhibitors of DPPIV. Modifications to the N terminus of Oxm modulated binding to the glucagon-like peptide (GLP)-1 receptor and degradation by DPPIV. Modifications to the midsection of Oxm modulated binding to the GLP-1 receptor and degradation by neutral endopeptidase. These modifications also altered bioactivity in vivo. The C-terminal octapeptide of Oxm was shown to contribute to the properties of Oxm in vitro and in vivo but was not alone sufficient for the effects of the peptide. Elongation and acylation of the C terminus of Oxm altered GLP-1 receptor binding and duration of action in vivo, which may be due to changes in peptide clearance. An Oxm analog was developed with enhanced pharmaceutical characteristics, with greater potency and longevity with respect to effects on food intake. These studies suggest that Oxm is a potential target for antiobesity drug design.


Subject(s)
Oxyntomodulin/chemistry , Oxyntomodulin/pharmacology , Structure-Activity Relationship , Amino Acid Sequence , Animals , Eating/drug effects , Glucagon-Like Peptide-1 Receptor , Male , Mice , Molecular Sequence Data , Oxyntomodulin/pharmacokinetics , Protein Binding , Rats , Receptors, Glucagon/metabolism , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...