Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
J Med Chem ; 63(11): 6134-6143, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32406685

ABSTRACT

Native insulin is susceptible to biophysical aggregation and fibril formation, promoted by manual agitation and elevated temperatures. The safety of the drug and its application to alternative forms of administration could be enhanced through the identification of chemical modifications that strengthen its physical stability without compromising its biological properties. Complex polysialic acids (PSAs) exist naturally and provide a means to enhance the physical properties of peptide therapeutics. A set of insulin analogues site-specifically derivatized with sialic acid were prepared in an overall yield of 50-60%. Addition of a single or multiple sialic acids conferred remarkable enhancement to the biophysical stability of human insulin while maintaining its potency. The time to the onset of fibrillation was extended by more than 10-fold relative to that of the native hormone. These results demonstrate that simplified sialic acid conjugates represent a viable alternative to complex natural PSAs in increasing the stability of therapeutic peptides.


Subject(s)
Insulin/analogs & derivatives , N-Acetylneuraminic Acid/chemistry , Animals , Blood Glucose/analysis , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/drug therapy , HEK293 Cells , Humans , Insulin/pharmacokinetics , Insulin/therapeutic use , Male , Mice , Mice, Inbred C57BL , Oligopeptides/chemical synthesis , Oligopeptides/chemistry , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Sialic Acids/chemistry , Therapeutic Equivalency
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.
Mol Microbiol ; 97(2): 229-43, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869931

ABSTRACT

The Phr peptides of the Bacillus species mediate quorum sensing, but their identification and function in other species of bacteria have not been determined. We have identified a Phr peptide quorum-sensing system (TprA/PhrA) that controls the expression of a lantibiotic gene cluster in the Gram-positive human pathogen, Streptococcus pneumoniae. Lantibiotics are highly modified peptides that are part of the bacteriocin family of antimicrobial peptides. We have characterized the basic mechanism for a Phr-peptide signaling system in S. pneumoniae and found that it induces the expression of the lantibiotic genes when pneumococcal cells are at high density in the presence of galactose, a main sugar of the human nasopharynx, a highly competitive microbial environment. Activity of the Phr peptide system is not seen when pneumococcal cells are grown with glucose, the preferred carbon source and the most prevalent sugar encountered by S. pneumoniae during invasive disease. Thus, the lantibiotic genes are expressed under the control of both cell density signals via the Phr peptide system and nutritional signals from the carbon source present, suggesting that quorum sensing and the lantibiotic machinery may help pneumococcal cells compete for space and resources during colonization of the nasopharynx.


Subject(s)
Bacteriocins/biosynthesis , Bacteriocins/genetics , Gene Expression Regulation, Bacterial , Multigene Family , Quorum Sensing/physiology , Streptococcus pneumoniae/genetics , Streptococcus pneumoniae/metabolism , Bacillus/genetics , Bacillus/metabolism , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Quorum Sensing/genetics
4.
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
5.
Mol Metab ; 3(3): 293-300, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24749059

ABSTRACT

For more than half a century glucagon has been used as a critical care medicine in the treatment of life-threatening hypoglycemia. It is commercially supplied as a lyophilized powder intended to be solubilized in dilute aqueous hydrochloric acid immediately prior to administration. We have envisioned a "ready-to-use" glucagon as a drug of more immediate and likely use. Through a series of iterative changes in the native sequence we have identified glucagon analogs of appreciably enhanced aqueous solubility at physiological pH, and of chemical stability suitable for routine medicinal use. The superior biophysical properties were achieved in part through adjustment of the isoelectric point by use of a C-terminal Asp-Glu dipeptide. The native glutamines at positions 3, 20 and 24 as well as the methionine at 27 were substituted with amino acids of enhanced chemical stability, as directed by a full alanine scan of the native hormone. Of utmost additional importance was the dramatically enhanced stability of the peptide when Ser16 was substituted with alpha,aminoisobutyric acid (Aib), a substitution that stabilizes peptide secondary structure. The collective set of changes yield glucagon analogs of comparable in vitro and in vivo biological character to native hormone but with biophysical properties much more suitable for clinical use.

6.
Int J Mass Spectrom ; 368: 6-14, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-26023288

ABSTRACT

Cross sections for 61 palmitoylated peptides and 73 cysteine-unmodified peptides are determined and used together with a previously obtained tryptic peptide library to derive a set of intrinsic size parameters (ISPs) for the palmitoyl (Pal) group (1.26 ± 0.04), carboxyamidomethyl (Am) group (0.92 ± 0.04), and the 20 amino acid residues to assess the influence of Pal- and Am-modification on cysteine and other amino acid residues. These values highlight the influence of the intrinsic hydrophobic and hydrophilic nature of these modifications on the overall cross sections. As a part of this analysis, we find that ISPs derived from a database of a modifier on one amino acid residue (CysPal) can be applied on the same modification group on different amino acid residues (SerPal and TyrPal). Using these ISP values, we are able to calculate peptide cross sections to within ± 2% of experimental values for 83% of Pal-modified peptide ions and 63% of Am-modified peptide ions. We propose that modification groups should be treated as individual contribution factors, instead of treating the combination of the particular group and the amino acid residue they are on as a whole when considering their effects on the peptide ion mobility features.

7.
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
8.
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
9.
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
10.
Am J Physiol Endocrinol Metab ; 305(2): E161-70, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23592482

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) plays a major role in promoting glucose-stimulated insulin secretion in pancreatic ß-cells. In the present study, we synthesized a novel functional analog of GLP-1 conjugated to tetramethyl rhodamine to monitor the internalization of the receptor. Our data show that after being internalized the receptor is sorted to lysosomes. In endosomes, receptor-ligand complex is found to be colocalized with adenylate cyclase. Pharmacological inhibition of endocytosis attenuates GLP-1R-mediated cAMP generation and consequent downstream protein kinase A substrate phosphorylation and glucose-stimulated insulin secretion. Our study underlines a paradigm shift in GLP-1R signaling and trafficking. The receptor ligand complex triggers cAMP generation both in plasma membrane and in endosomes, which has implications for receptor-mediated regulation of insulin secretion.


Subject(s)
Cyclic AMP/biosynthesis , Endosomes/metabolism , Glucose/pharmacology , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, Glucagon/physiology , Amino Acid Sequence , Blotting, Western , Cell Line , Exocytosis/physiology , Fluorescent Antibody Technique , Genes, Reporter , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Humans , Insulin Secretion , Insulin-Secreting Cells/drug effects , Luciferases/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Microscopy, Fluorescence , Molecular Sequence Data , Receptors, Glucagon/genetics , Sucrose/pharmacology
11.
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
12.
Biopolymers ; 98(5): 443-50, 2012.
Article in English | MEDLINE | ID: mdl-23203689

ABSTRACT

The ratio of GLP-1/glucagon receptor (GLP1R/GCGR) co-agonism that achieves maximal weight loss without evidence of hyperglycemia was determined in diet-induced obese (DIO) mice chronically treated with GLP1R/GCGR co-agonist peptides differing in their relative receptor agonism. Using glucagon-based peptides, a spectrum of receptor selectivity was achieved by a combination of selective incorporation of GLP-1 sequences, C-terminal modification, backbone lactam stapling to stabilize helical structure, and unnatural amino acid substitutions at the N-terminal dipeptide. In addition to α-amino-isobutyric acid (Aib) substitution at position two, we show that α,α'-dimethyl imidazole acetic acid (Dmia) can serve as a potent replacement for the highly conserved histidine at position one. Selective site-specific pegylation was used to further minimize enzymatic degradation and provide uniform, extended in vivo duration of action. Maximal weight loss devoid of any sign of hyperglycemia was achieved with a co-agonist comparably balanced for in vitro potency at murine GLP1R and GCGR. This peptide exhibited superior weight loss and glucose lowering compared to a structurally matched pure GLP1R agonist, and to co-agonists of relatively reduced GCGR tone. Any further enhancement of the relative GCGR agonist potency yielded increased weight loss but at the expense of elevated blood glucose. We conclude that GCGR agonism concomitant with GLP1R agonism constitutes a promising approach to treatment of the metabolic syndrome. However, the relative ratio of GLP1R/GCGR co-agonism needs to be carefully chosen for each species to maximize weight loss efficacy and minimize hyperglycemia.


Subject(s)
Glucagon-Like Peptide 1/agonists , Receptors, Glucagon/agonists , Weight Loss , Amino Acid Sequence , Amino Acid Substitution , Aminoisobutyric Acids/chemistry , Animals , Anti-Obesity Agents/chemical synthesis , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/standards , Blood Glucose/chemistry , Blood Glucose/drug effects , CHO Cells , Cricetinae , Cricetulus , Cyclic AMP/chemistry , Glucagon-Like Peptide 1/chemical synthesis , Glucagon-Like Peptide 1/pharmacokinetics , Glucagon-Like Peptide-1 Receptor , Glucose/adverse effects , Glucose/chemistry , Glucose/pharmacology , Glycogenolysis , Histidine/chemistry , Humans , Hyperglycemia/drug therapy , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , Proteolysis , Receptors, Glucagon/chemistry , Structure-Activity Relationship , Transfection
13.
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
14.
FASEB J ; 25(8): 2814-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21543764

ABSTRACT

Ghrelin is a hormone produced predominantly by the stomach that targets a number of specific areas in the central nervous system to promote a positive energy balance by increasing food intake and energy storage. In that respect, similarities exist with the effects of consuming a high-fat diet (HFD), which also increases caloric intake and the amount of stored calories. We determined whether the effects of ghrelin on feeding and adiposity are influenced by the exposure to an HFD. Chronic intracerebroventricular ghrelin (2.5 nmol/d) increased feeding in lean rats fed a low-fat control diet (CD) [192 ± 5 g (ghrelin+CD) vs. 152 ± 5 g (control i.c.v. saline+CD), P<0.001], but the combination of ghrelin plus HFD did not result in significantly greater hyperphagia [150 ± 7 g (ghrelin+HFD) vs. 136 ± 4 g (saline+HFD)]. Despite failing to increase food intake in rats fed the HFD, ghrelin nonetheless increased adiposity [fat mass increase of 14 ± 2 g (ghrelin+HFD) vs. 1 ± 1 g (saline+HFD), P<0.001] up-regulating the gene expression of lipogenic enzymes in white adipose tissue. Our findings demonstrate that factors associated with high-fat feeding functionally interact with pathways regulating the effect of ghrelin on food intake. We conclude that ghrelin's central effects on nutrient intake and nutrient partitioning can be separated and suggest an opportunity to identify respective independent neuronal pathways.


Subject(s)
Adiposity/drug effects , Ghrelin/pharmacology , Adipose Tissue, White/drug effects , Adipose Tissue, White/physiology , Adiposity/physiology , Animals , Dietary Fats/administration & dosage , Eating/drug effects , Eating/physiology , Ghrelin/administration & dosage , Ghrelin/physiology , Hyperphagia/etiology , Hyperphagia/physiopathology , Hypothalamus, Middle/drug effects , Hypothalamus, Middle/physiology , Infusions, Intraventricular , Lipogenesis/drug effects , Lipogenesis/genetics , Lipogenesis/physiology , Male , Melanocortins/antagonists & inhibitors , Melanocortins/physiology , Neuropeptides/physiology , Rats , Rats, Long-Evans , Rats, Wistar , Receptors, Neuropeptide/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation
15.
ACS Chem Biol ; 6(2): 135-45, 2011 Feb 18.
Article in English | MEDLINE | ID: mdl-20939591

ABSTRACT

Ex-4 (9-39)a is a well characterized GLP-1 receptor antagonist that suffers from two notable limitations, its nonhuman amino acid sequence and its relatively short in vivo duration of action. Comparable N-terminal shortening of human GLP-1 lessens agonism but does not provide a high potency antagonist. Through a series of GLP-1/Ex-4 hybrid peptides, the minimal structural changes required to generate a pure GLP-1-based antagonist were identified as Glu16, Val19, and Arg20, yielding an antagonist of approximately 3-fold greater in vitro potency compared with Ex-4 (9-39)a. The structural basis of antagonism appears to result from stabilization of the α helix combined with enhanced electrostatic and hydrophobic interactions with the extracellular domain of the receptor. Site-specific acylation of the human-based antagonist yielded a peptide of increased potency as a GLP-1 receptor antagonist and 10-fold greater selectivity relative to the GIP receptor. The acylated antagonist demonstrated sufficient duration of action to maintain inhibitory activity when administered as a daily subcutaneous injection. The sustained pharmacokinetics and enhanced human sequence combine to form an antagonist optimized for clinical study. Daily administration of this antagonist by subcutaneous injection to diet-induced obese mice for 1 week caused a significant increase in food intake, body weight, and glucose intolerance, demonstrating endogenous GLP-1 as a relevant hormone in mammalian energy balance in the obese state.


Subject(s)
Glucagon-Like Peptide 1/metabolism , Receptors, Glucagon/antagonists & inhibitors , Acylation , Amino Acid Sequence , Animals , Binding Sites , Body Weight/drug effects , Dietary Fats/administration & dosage , Eating/drug effects , Energy Metabolism/drug effects , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/chemistry , Glucagon-Like Peptide 1/pharmacokinetics , Humans , Mice , Mice, Obese , Molecular Sequence Data , Obesity/chemically induced , Obesity/drug therapy , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacokinetics , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism
16.
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
17.
Protein Pept Lett ; 15(2): 232-4, 2008.
Article in English | MEDLINE | ID: mdl-18289117

ABSTRACT

Oxyntomodulin is a proglucagon-derived gut hormone that reduces food intake and body weight, thus represents a potential therapy for obesity. We synthesized and crystallized oxyntomodulin. The crystal diffracts x-ray to 2.4 A resolution and belongs to space group P2(1)3 with unit-cell parameters a=b=c= 48.44 A, alpha=beta=gamma=90 degrees . Preliminary analysis indicates a trimer packing in one asymmetric unit.


Subject(s)
Anti-Obesity Agents/chemistry , Oxyntomodulin/chemistry , X-Ray Diffraction , Animals , Body Weight , Crystallization , Oxyntomodulin/chemical synthesis
18.
Acta Crystallogr Sect F Struct Biol Cryst Commun ; 63(Pt 7): 599-601, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17620721

ABSTRACT

Glucagon and glucagon-like peptide 1 (GLP-1) are drugs or drug candidates for the treatment of metabolic diseases such as diabetes and obesity. The native hormones have pharmacological deficiencies such as short half-life and poor solubility. A novel glucagon receptor agonist named glucagon-Cex has been designed, synthesized and crystallized. This peptide was highly soluble under physiological conditions and crystallized readily. The crystal diffracted X-rays to 2.2 A resolution and the diffraction was consistent with space group P23, with unit-cell parameters a = b = c = 48.20 A, alpha = beta = gamma = 90.0 degrees. The crystals were suitable for a full structural determination to reveal the conformational differences between glucagon-Cex and the native hormone.


Subject(s)
Glucagon/chemical synthesis , Glucagon/therapeutic use , Crystallization , Crystallography, X-Ray , Glucagon/analogs & derivatives , Obesity/drug therapy , Peptide Fragments/chemical synthesis , Peptide Fragments/therapeutic use
19.
Endocrinology ; 146(12): 5257-66, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16166226

ABSTRACT

alphaMSH has generally been accepted as the endogenous ligand for melanocortin 4 receptor (MC4R), which plays a major role in energy homeostasis. Targeting MC4R to develop antiobesity agents, many investigators have performed a structure-activity relationship (SAR) studies based on alphaMSH structure. In this report, we performed a SAR study using human betaMSH (5 - 22) (DEGPYRMEHFRWGSPPKD, peptide 1) as a lead sequence to develop potent and selective agonists for MC4R and MC3R. The SAR study was begun with a truncation of N terminus of betaMSH (5 - 22) together with acetylation of the N terminus and amidation of the C terminus of the peptide. Introduction of a cyclic disulfide constrain and replacement of L-Phe with D-Phe afforded a super potent agonist (peptide 5). Furthermore truncation at the C terminus generated a small and potent MC4R and MC3R agonist (Ac-YRcyclo[CEHdFRWC]amide, peptide 6), which exhibited no MC5R and greatly reduced MC1R activity. Molecular modeling of Ac-YRcyclo[CEHdFRWC]amide (peptide 6) revealed that Arg2 in the peptide formed a salt bridge with Glu4. Subcutaneous or intracerebroventricular administration of peptide 6 in rats showed potent in vivo efficacy as evidenced by its effects in reducing energy balance, increasing fat use, and decreasing weight gain in both acute and chronic rat metabolic studies. Furthermore, the antiobesity effect by peptide 6 was manifested only in wild-type but not MC4R-deficient mice, indicating that antiobesity effects of the peptide were attributed largely through MC4R but not MC3R agonist activity of the peptide.


Subject(s)
Diet , Eating/drug effects , Melanocyte-Stimulating Hormones/pharmacology , Obesity/physiopathology , Peptide Fragments/pharmacology , Receptor, Melanocortin, Type 4/agonists , Weight Gain/drug effects , Animals , Body Composition , Body Weight , Dose-Response Relationship, Drug , Energy Metabolism , Injections, Intraventricular , Injections, Subcutaneous , Male , Melanocyte-Stimulating Hormones/chemistry , Models, Molecular , Molecular Structure , Obesity/etiology , Obesity/pathology , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Rats , Rats, Long-Evans , Structure-Activity Relationship
20.
Bioorg Med Chem Lett ; 15(20): 4611-4, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16105738

ABSTRACT

Extensive structure-activity relationship studies utilizing a beta-MSH-derived cyclic nonapeptide, Ac-Tyr-Arg-[Cys-Glu-His-D-Phe-Arg-Trp-Cys]-NH(2) (3), led to identification of a series of novel MC-4R selective disulfide-constrained hexapeptide analogs including Ac-[hCys-His-D-Phe-Arg-Trp-Cys]-NH(2) (12). The structural modifications associated with profound influence on MC-4R potency and selectivity were ring size, ring conformation, and the aromatic substitution of the D-Phe7. These cyclic peptide analogs provide novel and enhanced reagents for use in the elucidation of melanocortin-4 receptor-related physiology, and may additionally find application in the treatment of obesity and related metabolic disorders.


Subject(s)
Disulfides/chemistry , Receptor, Melanocortin, Type 4/agonists , Chromatography, High Pressure Liquid , Mass Spectrometry , Receptor, Melanocortin, Type 4/chemistry , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL
...