Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 172
Filter
1.
Mol Metab ; 46: 101090, 2021 04.
Article in English | MEDLINE | ID: mdl-32987188

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 receptor (GLP-1R) agonists are approved to treat type 2 diabetes and obesity. They elicit robust improvements in glycemic control and weight loss, combined with cardioprotection in individuals at risk of or with pre-existing cardiovascular disease. These attributes make GLP-1 a preferred partner for next-generation therapies exhibiting improved efficacy yet retaining safety to treat diabetes, obesity, non-alcoholic steatohepatitis, and related cardiometabolic disorders. The available clinical data demonstrate that the best GLP-1R agonists are not yet competitive with bariatric surgery, emphasizing the need to further improve the efficacy of current medical therapy. SCOPE OF REVIEW: In this article, we discuss data highlighting the physiological and pharmacological attributes of potential peptide and non-peptide partners, exemplified by amylin, glucose-dependent insulinotropic polypeptide (GIP), and steroid hormones. We review the progress, limitations, and future considerations for translating findings from preclinical experiments to competitive efficacy and safety in humans with type 2 diabetes and obesity. MAJOR CONCLUSIONS: Multiple co-agonist combinations exhibit promising clinical efficacy, notably tirzepatide and investigational amylin combinations. Simultaneously, increasing doses of GLP-1R agonists such as semaglutide produces substantial weight loss, raising the bar for the development of new unimolecular co-agonists. Collectively, the available data suggest that new co-agonists with robust efficacy should prove superior to GLP-1R agonists alone to treat metabolic disorders.


Subject(s)
Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Metabolic Diseases/drug therapy , Adipose Tissue , Animals , Cardiovascular Diseases , Diabetes Mellitus, Type 2/metabolism , Fatty Liver , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Obesity/metabolism , Receptors, Gastrointestinal Hormone/drug effects , Weight Loss
2.
Life Sci ; 257: 118025, 2020 Sep 15.
Article in English | MEDLINE | ID: mdl-32598933

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 receptor (GLP-1R) and glucose-dependent insulinotropic polypeptide receptor (GIPR) co-agonists have emerged as treatment options for reversing diabetes and obesity. Here, we screened the high potency receptor-biased GLP-1R agonists via a newly designed high-throughput GLP-1R extracellular domain (ECD)-based system and demonstrated its in vitro and in vivo therapeutic characters. METHODS: Twelve 9-mer peptides (named XEL1-XEL12) which were screened from a large phage-displayed peptide library were fused to the N-terminus of GIP (3-30) to generate another twelve fusion peptides, termed XEL13-24. Using the six lysine-altered XEL17 as leading sequences, eighteen fatty chain modified fusion peptides were further assessed via in vitro GLP-1R/GIPR-based cell assay. Moreover, the acute and long-acting in vivo effects of selected candidate on diabetic db/db mice and diet-induced obesity (DIO) rats were both carefully evaluated. RESULTS: XEL17 exhibited balanced activation potency on GLP-1R/GIPR in stable cell lines, and further assessment was performed to evaluate the XEL32, a fatty chain modified XEL17 derivative. Preclinical pharmacodynamic results in diabetic db/db mice demonstrated that XEL32 held outstanding insulinotropic and glucose-lowering activities. In addition, protracted antidiabetic effects of XEL32 were also proved by the hypoglycemic test and multiple oral glucose tolerance test. Furthermore, chronic treatment of XEL32 in DIO rats exhibited outstanding beneficial effects on body weight control, fat loss, food intake control, hemoglobin A1C (HbA1C) reduction as well as the glucose tolerance. CONCLUSIONS: XEL32, as a novel GLP-1/GIP dual receptor agonist, may supply efficient glycemic control and weight loss.


Subject(s)
Glucagon-Like Peptide-1 Receptor/metabolism , Peptides/pharmacology , Receptors, Gastrointestinal Hormone/metabolism , Weight Loss/drug effects , Animals , Blood Glucose/metabolism , China , Diabetes Mellitus/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/drug effects , Glucose Tolerance Test , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Hypoglycemic Agents/pharmacology , Male , Mice , Obesity/metabolism , Rats , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/drug effects
3.
Curr Opin Pharmacol ; 43: 118-123, 2018 12.
Article in English | MEDLINE | ID: mdl-30273889

ABSTRACT

Dumping syndrome is a common and debilitating complication of upper gastrointestinal surgery. Accelerated gastric emptying and dysregulated secretion of gastrointestinal (GI) hormones are involved in its pathophysiology. Pasireotide, a novel somatostatin analogue, improved dumping in a phase-2 study. Preliminary data suggest that the glucagon-like peptide-1 (GLP-1) analogue liraglutide can also improve dumping. Short bowel syndrome is the most common cause of intestinal failure and involves not only a loss of mucosal absorptive area but also hypersecretion and accelerated transit. GLP-2 is the best studied hormone involved in intestinal adaptation. An increasing body of evidence demonstrates that the GLP-2 analogue teduglutide reduces parenteral support needs. New GLP-2 analogues and analogues of other GI hormones such as liraglutide are being investigated as promising treatments in short bowel syndrome.


Subject(s)
Dumping Syndrome/drug therapy , Gastrointestinal Agents/therapeutic use , Gastrointestinal Motility/drug effects , Gastrointestinal Tract/drug effects , Intestinal Absorption/drug effects , Receptors, Gastrointestinal Hormone/drug effects , Short Bowel Syndrome/drug therapy , Animals , Dumping Syndrome/metabolism , Dumping Syndrome/physiopathology , Gastrointestinal Agents/adverse effects , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiopathology , Humans , Ligands , Liraglutide/therapeutic use , Peptides/therapeutic use , Receptors, Gastrointestinal Hormone/metabolism , Short Bowel Syndrome/metabolism , Short Bowel Syndrome/physiopathology , Signal Transduction/drug effects , Somatostatin/analogs & derivatives , Somatostatin/therapeutic use , Treatment Outcome
4.
Endocr Dev ; 32: 165-182, 2017.
Article in English | MEDLINE | ID: mdl-28873389

ABSTRACT

Obesity and its comorbidities such as type 2 diabetes constitute major worldwide health threats, and the identification of an effective medical intervention has emerged as a global priority. The limited effectiveness of historical, anti-obesity treatments is commonly attributed to the complexity of the disease and the redundancy of metabolic regulatory mechanisms that sustain body weight. At the forefront of obesity research is the development of combinational drug therapies that simultaneously target multiple regulatory pathways, which promote dysfunctional metabolism. Recently, molecularly crafted unimolecular "multi-agonism" of balanced activity at 3 key receptors involved in metabolism and specifically the glucagon-like peptide (GLP)-1 receptor, glucose-dependent insulinotropic polypeptide (GIP) receptor and glucagon receptor was reported as superior to conventional monoagonist therapy. These mixed peptide agonists are designed to pharmacologically integrate the insulinotropic and anorexigenic effects of GLP-1, the thermogenic and lipolytic activities of glucagon, and the insulinotropic and insulin sensitizing properties of GIP. The molecular mechanism of these purposefully promiscuous ligands is not completely understood, however, recent studies in pancreatic beta cells point to the prospect of a complex signaling network that can magnify the signaling of multi-agonist ligands. The activation of this signalosome might explain the additional therapeutic benefit inherent to simultaneous cellular activation through multiple metabolic receptors.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Gastrointestinal Hormones/physiology , Gastrointestinal Microbiome/physiology , Neuroendocrine Cells/physiology , Obesity/therapy , Peptide Hormones/agonists , Animals , Body Weight , Germ-Free Life/physiology , Humans , Insulin-Secreting Cells/metabolism , Mice , Peptide Hormones/physiology , Prebiotics/administration & dosage , Receptor Cross-Talk , Receptors, Gastrointestinal Hormone/drug effects
5.
Nat Rev Gastroenterol Hepatol ; 13(1): 38-48, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26392067

ABSTRACT

The gastrointestinal tract is the major source of the related hormones ghrelin and motilin, which act on structurally similar G protein-coupled receptors. Nevertheless, selective receptor agonists are available. The primary roles of endogenous ghrelin and motilin in the digestive system are to increase appetite or hedonic eating (ghrelin) and initiate phase III of gastric migrating myoelectric complexes (motilin). Ghrelin and motilin also both inhibit nausea. In clinical trials, the motilin receptor agonist camicinal increased gastric emptying, but at lower doses reduced gastroparesis symptoms and improved appetite. Ghrelin receptor agonists have been trialled for the treatment of diabetic gastroparesis because of their ability to increase gastric emptying, but with mixed results; however, relamorelin, a ghrelin agonist, reduced nausea and vomiting in patients with this disorder. Treatment of postoperative ileus with a ghrelin receptor agonist proved unsuccessful. Centrally penetrant ghrelin receptor agonists stimulate defecation in animals and humans, although ghrelin itself does not seem to control colorectal function. Thus, the most promising uses of motilin receptor agonists are the treatment of gastroparesis or conditions with slow gastric emptying, and ghrelin receptor agonists hold potential for the reduction of nausea and vomiting, and the treatment of constipation. Therapeutic, gastrointestinal roles for receptor antagonists or inverse agonists have not been identified.


Subject(s)
Gastrointestinal Agents/therapeutic use , Gastrointestinal Diseases/drug therapy , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Ghrelin/drug effects , Receptors, Neuropeptide/drug effects , Appetite/drug effects , Appetite/physiology , Constipation/drug therapy , Constipation/physiopathology , Gastric Emptying/drug effects , Gastric Emptying/physiology , Gastrointestinal Agents/adverse effects , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Ghrelin/physiology , Humans , Hunger/drug effects , Hunger/physiology , Motilin/physiology , Nausea/drug therapy , Nausea/physiopathology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/physiology , Receptors, Ghrelin/agonists , Receptors, Ghrelin/physiology , Receptors, Neuropeptide/agonists , Receptors, Neuropeptide/physiology , Signal Transduction/physiology
6.
Am J Physiol Gastrointest Liver Physiol ; 307(11): G1108-14, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25324507

ABSTRACT

Animal studies have demonstrated that glucose-dependent insulinotropic polypeptide (GIP) and GIP receptor (GIPR) contribute to the etiology of obesity. In humans, genomewide association studies have identified single nucleotide polymorphisms (SNPs) in the GIPR gene that are strongly associated with body mass index (BMI); however, it is not clear whether genetic variations in the GIP gene are involved in the development of obesity. In the current study, we assessed the impact of GIP SNPs on obesity-related traits in Japanese adults. Six tag SNPs were tested for associations with obesity-related traits in 3,013 individuals. Multiple linear regression analyses showed that rs9904288, located at the 3'-end of GIP, was significantly associated with visceral fat area (VFA). Moreover, rs1390154 and rs4794008 showed significant associations with plasma triglyceride levels and hemoglobin A1c levels, respectively. Among the significant SNPs, rs9904288 and rs1390154 were independently linked with SNPs in active enhancers of the duodenum mucosa, the main GIP-secreting tissue. The haplotypes of these two SNPs exhibited stronger associations with VFA. Numbers of VFA-increasing alleles of rs9904288 and BMI-increasing alleles of previously identified GIPR SNPs showed a strong additive effect on VFA, waist circumference, and BMI in the subject population. These novel results support the notion that the GIP-GIPR axis plays a role in the etiology of central obesity in humans, which is characterized by the accumulation of visceral fat.


Subject(s)
Abdominal Fat/physiology , Gastric Inhibitory Polypeptide/genetics , Obesity/genetics , Alleles , Female , Genome-Wide Association Study , Haplotypes , Humans , Japan/epidemiology , Male , Middle Aged , Obesity/epidemiology , Polymorphism, Single Nucleotide/genetics , Receptors, Gastrointestinal Hormone/drug effects
7.
Br J Pharmacol ; 171(13): 3132-45, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24628305

ABSTRACT

Class B GPCRs of the secretin family are important drug targets in many human diseases including diabetes, neurodegeneration, cardiovascular disease and psychiatric disorders. X-ray crystal structures for the glucagon receptor and corticotropin-releasing factor receptor 1 have now been published. In this review, we analyse the new structures and how they compare with each other and with Class A and F receptors. We also consider the differences in druggability and possible similarity in the activation mechanisms. Finally, we discuss the potential for the design of small-molecule modulators for these important targets in drug discovery. This new structural insight allows, for the first time, structure-based drug design methods to be applied to Class B GPCRs.


Subject(s)
Drug Design , Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/chemistry , Animals , Crystallography, X-Ray , Drug Discovery/methods , Humans , Molecular Targeted Therapy , Protein Conformation , Receptors, Corticotropin-Releasing Hormone/chemistry , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/chemistry
8.
Neuropeptides ; 47(2): 59-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23352609

ABSTRACT

Short selective neuropeptide Y (NPY) analogs are highly attractive because of their facile synthesis. Based on the reduced-size NPY analog [Pro(30), Nle(31), Bpa(32), Leu(34)]NPY 28-36 position 32 was identified as a key position to alter the preferential activation pattern of the human neuropeptide Y receptors (hYRs). By replacing benzoylphenylalanine (Bpa) by a biphenylalanine (Bip) the photostability was first improved while the biological activity was maintained. SAR-studies showed that both aromatic rings have a high influence on the preferential hYR subtype activation. Interestingly, replacement of Bpa(32) by a strongly hydrophobic moiety changed the hYR subtype preference of the analog. Whereas the parent compound is able to activate the human neuropeptide Y1 receptor (hY1R) subtype, the introduction of an N(ε)-ortho-carbaboranyl propionic acid modified lysine resulted in a loss of activity at the hY1R but in an increased activity at both the hY2R and the hY4R. However, subsequent receptor internalization studies with this novel analog revealed that receptor internalization can neither be triggered at the hY2R nor at the hY4R suggesting a biased ligand. Surprisingly, investigations by (1)H NMR spectroscopy revealed structural changes in the side chains of residues Pro(30) and Leu(34) which nicely correlates with the shift from hY1R/hY4R to hY2R/hY4R activation preference. Thus, position 32 has been identified to switch the bioactive conformation and subsequently influences receptor subtype activation behavior.


Subject(s)
Boranes/pharmacology , Neuropeptide Y/analogs & derivatives , Neuropeptide Y/pharmacology , Receptors, Gastrointestinal Hormone/drug effects , Amino Acid Sequence , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , HEK293 Cells , Humans , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Neuropeptide Y/chemical synthesis , Protein Conformation , Structure-Activity Relationship
9.
Diabetes ; 59(6): 1445-50, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20332343

ABSTRACT

OBJECTIVE: We previously showed that peroxisome proliferator-activated receptor (PPAR)-gamma in beta-cells regulates pdx-1 transcription through a functional PPAR response element (PPRE). Gene Bank blast for a homologous nucleotide sequence revealed the same PPRE within the rat glucose-dependent insulinotropic polypeptide receptor (GIP-R) promoter sequence. We investigated the role of PPARgamma in GIP-R transcription. RESEARCH DESIGN AND METHODS: Chromatin immunoprecipitation assay, siRNA, and luciferase gene transcription assay in INS-1 cells were performed. Islet GIP-R expression and immunohistochemistry studies were performed in pancreas-specific PPARgamma knockout mice (PANC PPARgamma(-/-)), normoglycemic 60% pancreatectomy rats (Px), normoglycemic and hyperglycemic Zucker fatty (ZF) rats, and mouse islets incubated with troglitazone. RESULTS: In vitro studies of INS-1 cells confirmed that PPAR-gamma binds to the putative PPRE sequence and regulates GIP-R transcription. In vivo verification was shown by a 70% reduction in GIP-R protein expression in islets from PANC PPARgamma(-/-) mice and a twofold increase in islets of 14-day post-60% Px Sprague-Dawley rats that hyperexpress beta-cell PPARgamma. Thiazolidinedione activation (72 h) of this pathway in normal mouse islets caused a threefold increase of GIP-R protein and a doubling of insulin secretion to 16.7 mmol/l glucose/10 nmol/l GIP. Islets from obese normoglycemic ZF rats had twofold increased PPARgamma and GIP-R protein levels versus lean rats, with both lowered by two-thirds in ZF rats made hyperglycemic by 60% Px. CONCLUSIONS: Our studies have shown physiologic and pharmacologic regulation of GIP-R expression in beta-cells by PPARgamma signaling. Also disruption of this signaling pathway may account for the lowered beta-cell GIP-R expression and resulting GIP resistance in type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Insulin-Secreting Cells/physiology , PPAR gamma/physiology , Receptors, Gastrointestinal Hormone/genetics , Animals , Chromans/pharmacology , Drug Resistance , Gastric Inhibitory Polypeptide/physiology , Glucose/physiology , Male , Mice , PPAR gamma/deficiency , PPAR gamma/genetics , Pancreatectomy , Rats , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Gastrointestinal Hormone/physiology , Thiazolidinediones/pharmacology , Troglitazone
10.
Pharmacol Ther ; 125(1): 39-54, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19686775

ABSTRACT

Recent research and clinical data have begun to demonstrate the huge potential therapeutic importance of ligands that modulate the activity of the secretin-like, Class II, G protein-coupled receptors (GPCRs). Ligands that can modulate the activity of these Class II GPCRs may have important clinical roles in the treatment of a wide variety of conditions such as osteoporosis, diabetes, amyotrophic lateral sclerosis and autism spectrum disorders. While these receptors present important new therapeutic targets, the large glycoprotein nature of their cognate ligands poses many problems with respect to therapeutic peptidergic drug design. These native peptides often exhibit poor bioavailability, metabolic instability, poor receptor selectivity and resultant low potencies in vivo. Recently, increased attention has been paid to the structural modification of these peptides to enhance their therapeutic efficacy. Successful modification strategies have included d-amino acid substitutions, selective truncation, and fatty acid acylation of the peptide. Through these and other processes, these novel peptide ligand analogs can demonstrate enhanced receptor subtype selectivity, directed signal transduction pathway activation, resistance to proteolytic degradation, and improved systemic bioavailability. In the future, it is likely, through additional modification strategies such as addition of circulation-stabilizing transferrin moieties, that the therapeutic pharmacopeia of drugs targeted towards Class II secretin-like receptors may rival that of the Class I rhodopsin-like receptors that currently provide the majority of clinically used GPCR-based therapeutics. Currently, Class II-based drugs include synthesized analogs of vasoactive intestinal peptide for type 2 diabetes or parathyroid hormone for osteoporosis.


Subject(s)
Drug Design , Neurosecretory Systems/drug effects , Neurotransmitter Agents/pharmacology , Peptide Hormones/pharmacology , Receptors, G-Protein-Coupled/drug effects , Amino Acid Sequence , Animals , Humans , Ligands , Molecular Sequence Data , Molecular Structure , Neurosecretory Systems/metabolism , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/pharmacokinetics , Neurotransmitter Agents/therapeutic use , Peptide Hormones/chemistry , Peptide Hormones/pharmacokinetics , Peptide Hormones/therapeutic use , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Gastrointestinal Hormone/metabolism , Structure-Activity Relationship
11.
J Physiol ; 587(1): 19-25, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19064614

ABSTRACT

The last decade has witnessed a marked increase in our understanding of the importance of gut hormones in the regulation of energy homeostasis. In particular, the discovery that the gut hormone peptide YY 3-36 (PYY3-36) reduced feeding in obese rodents and humans fuelled interest in the role of PYY3-36 in body weight regulation. Pharmacological and genetic approaches have revealed that the Y2-receptor mediates the anorectic effects of PYY3-36 whilst mechanistic studies in rodents identified the hypothalamus, vagus and brainstem regions as potential sites of action. More recently, using functional brain imaging techniques in humans, PYY3-36 was found to modulate neuronal activity within hypothalamic and brainstem, and brain regions involved in reward processing. Several lines of evidence suggest that low circulating PYY concentrations predispose towards the development and or maintenance of obesity. Subjects with reduced postprandial PYY release exhibit lower satiety and circulating PYY levels that correlate negatively with markers of adiposity. In addition, mice lacking PYY are hyperphagic and become obese. Conversely, chronic PYY3-36 administration to obese rodents reduces adiposity, and transgenic mice with increased circulating PYY are resistant to diet-induced obesity. Moreover, there is emerging evidence that PYY3-36 may partly mediate the reduced appetite and weight loss benefits observed post-gastric bypass surgery. Taken together these findings, coupled with the retained responsiveness of obese subjects to the effects of PYY3-36, suggest that targeting the PYY system may offer a therapeutic strategy to help treat obesity.


Subject(s)
Appetite Regulation/physiology , Obesity/physiopathology , Peptide YY/physiology , Animals , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Gastric Bypass , Humans , Obesity/drug therapy , Obesity/surgery , Peptide Fragments , Peptide YY/administration & dosage , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Gastrointestinal Hormone/physiology , Weight Loss/physiology
14.
Peptides ; 29(8): 1390-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18440094

ABSTRACT

Although the neuropeptide Y (NPY) family has been demonstrated to control bone metabolism, the role of pancreatic polypeptide (PP), which has structural homology with NPY and peptide YY (PYY) to share the NPY family receptors, in peripheral bone tissues has remained unknown. In the present study, we studied the regulatory roles of PP and its Y receptors using MC3T3-E1 cells, a murine transformed osteoblastic cell line, as a model for osteoblastic differentiation. We found that (1) PP mRNA was detected and increased during cell-contact-induced differentiation in MC3T3-E1 cells; (2) the immunoreactivity of PP was detected by radioimmunoassay and increased in culture medium during differentiation; (3) all the types of NPY family receptor mRNAs (Y1, Y2, Y4, Y5, and y6) were found to increase during differentiation; (4) PP stimulated differentiation in MC3T3-E1 cells in terms of ALP mRNA and BMP-2 mRNA. These findings suggested that MC3T3-E1 cells produce and secrete PP, which may in turn stimulate the differentiation of MC3T3-E1 through its specific receptors in an autocrine manner.


Subject(s)
Cell Differentiation/physiology , Osteoblasts/metabolism , Pancreatic Polypeptide/metabolism , Pancreatic Polypeptide/physiology , Receptors, Gastrointestinal Hormone/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Mice , Osteoblasts/cytology , Pancreatic Polypeptide/genetics , RNA, Messenger/biosynthesis , Receptors, Gastrointestinal Hormone/drug effects , Reverse Transcriptase Polymerase Chain Reaction
16.
Diabetes ; 56(12): 3006-13, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17717280

ABSTRACT

OBJECTIVE: Dipeptidyl peptidase-4 (DPP4) inhibitors lower blood glucose in diabetic subjects; however, the mechanism of action through which these agents improve glucose homeostasis remains incompletely understood. Although glucagon-like peptide (GLP)-1 and glucose-dependent insulinotropic polypeptide (GIP) represent important targets for DPP4 activity, whether additional substrates are important for the glucose-lowering actions of DPP4 inhibitors remains uncertain. RESEARCH DESIGN AND METHODS: We examined the efficacy of continuous vildagliptin administration in wild-type (WT) and dual incretin receptor knockout (DIRKO) mice after 8 weeks of a high-fat diet. RESULTS: Vildagliptin had no significant effect on food intake, energy expenditure, body composition, body weight gain, or insulin sensitivity in WT or DIRKO mice. However, glycemic excursion after oral glucose challenge was significantly reduced in WT but not in DIRKO mice after vildagliptin treatment. Moreover, vildagliptin increased levels of glucose-stimulated plasma insulin and reduced levels of cholesterol and triglycerides in WT but not in DIRKO mice. Vildagliptin treatment reduced the hepatic expression of genes important for cholesterol synthesis and fatty acid oxidation, including phospho-mevalonate kinase (Mvk), acyl-coenzyme dehydrogenase medium chain (Acadm), mevalonate (diphospho)decarboxylase (Mvd), and Acyl-CoA synthetase (Acsl1), in WT but not in DIRKO mice. However, vildagliptin also reduced levels of hepatic mRNA transcripts for farnesyl di-phosphate transferase (Fdft1), acetyl coenzyme A acyltransferase 1 (Acaa1), and carnitine palmitoyl transferase 1 (Cpt 1) in DIRKO mice. No direct effect of GLP-1 receptor agonists was detected on cholesterol or triglyceride synthesis and secretion in WT hepatocytes. CONCLUSIONS: These findings illustrate that although GLP-1 and GIP receptors represent the dominant molecular mechanisms for transducing the glucoregulatory actions of DPP4 inhibitors, prolonged DPP4 inhibition modulates the expression of genes important for lipid metabolism independent of incretin receptor action in vivo.


Subject(s)
Adamantane/analogs & derivatives , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/physiology , Nitriles/therapeutic use , Pyrrolidines/therapeutic use , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Receptors, Peptide/physiology , Adamantane/therapeutic use , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cell Division/drug effects , Glucagon-Like Peptide-1 Receptor , Glucose Tolerance Test , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/physiology , Mice , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Glucagon/drug effects , Receptors, Peptide/deficiency , Receptors, Peptide/genetics , Vildagliptin
17.
Inflammopharmacology ; 15(1): 36-42, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17323194

ABSTRACT

The prokinetic effects of mitemcinal, an orally active motilin receptor agonist, on the lower gastrointestinal tracts were investigated in conscious dogs. Oral administration of mitemcinal (0.1-1 mg/kg) stimulated colonic motility, which was measured by chronically implanted force-transducers, as well as gastric motility in a dose-dependent manner. The gastrointestinal contractile activities induced by mitemcinal were inhibited by the continuous intravenous infusion of GM-109, a selective motilin receptor antagonist. Oral administration of mitemcinal (0.3-3 mg/kg) also accelerated bowel movement after feeding without inducing diarrhea in dogs. The results demonstrate that mitemcinal stimulates colonic motility via motilin receptors and the effect of mitemcinal on colonic motility may reflect bowel movement after feeding. Thus, mitemcinal could be a promising agent for treatment of not only the upper but also the lower gastrointestinal motility disorders.


Subject(s)
Colon/drug effects , Defecation/drug effects , Erythromycin/analogs & derivatives , Gastrointestinal Motility/drug effects , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Neuropeptide/drug effects , Administration, Oral , Animals , Colon/physiology , Diarrhea , Dogs , Dose-Response Relationship, Drug , Erythromycin/administration & dosage , Erythromycin/pharmacology , Female , Male , Motilin/analogs & derivatives , Motilin/pharmacology , Muscle Contraction/drug effects , Receptors, Gastrointestinal Hormone/agonists , Receptors, Neuropeptide/agonists
18.
Biol Chem ; 388(2): 221-6, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17261085

ABSTRACT

Effects of chemical ablation of the GIP and GLP-1 receptors on metabolic aspects of obesity-diabetes were investigated using the stable receptor antagonists (Pro3)GIP and exendin(9-39)amide. Ob/ob mice received a daily i.p. injection of saline vehicle, (Pro3)GIP, exendin(9-39)amide or a combination of both peptides over a 14-day period. Non-fasting plasma glucose levels were significantly (p<0.05) lower in (Pro3)GIP-treated mice compared to control mice after just 9 days of treatment. (Pro3)GIP-treated mice also displayed significantly lower plasma glucose concentrations in response to feeding and intraperitoneal administration of either glucose or insulin (p<0.05 to p<0.001). The (Pro3)GIP-treated group also exhibited significantly (p<0.05) reduced pancreatic insulin content. Acute administration of exendin(9-39)amide immediately prior to re-feeding completely annulled the beneficial effects of sub-chronic (Pro3)GIP treatment, but non-fasting concentrations of active GLP-1 were unchanged. Combined sub-chronic administration of (Pro3GIP) with exendin(9-39)amide revealed no beneficial effects. Similarly, daily administration of exendin(9-39)amide alone had no significant effects on any of the metabolic parameters measured. These studies highlight an important role for GIP in obesity-related forms of diabetes, suggesting the possible involvement of GLP-1 in the beneficial actions of GIP receptor antagonism.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Gastric Inhibitory Polypeptide/administration & dosage , Obesity/metabolism , Peptide Fragments/administration & dosage , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Glucagon/metabolism , Animals , Blood Glucose/chemistry , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Disease Models, Animal , Dose-Response Relationship, Drug , Glucagon-Like Peptide-1 Receptor , Injections, Intraperitoneal , Insulin/administration & dosage , Insulin/chemistry , Insulin/metabolism , Mice , Mice, Obese , Obesity/complications , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/drug effects , Receptors, Glucagon/chemistry , Receptors, Glucagon/drug effects , Structure-Activity Relationship , Time Factors
20.
Diabetes ; 54(8): 2436-46, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16046312

ABSTRACT

Glucose-dependent insulinotropic polypeptide (gastric inhibitory polypeptide [GIP]) is an important incretin hormone secreted by endocrine K-cells in response to nutrient ingestion. In this study, we investigated the effects of chemical ablation of GIP receptor (GIP-R) action on aspects of obesity-related diabetes using a stable and specific GIP-R antagonist, (Pro3)GIP. Young adult ob/ob mice received once-daily intraperitoneal injections of saline vehicle or (Pro3)GIP over an 11-day period. Nonfasting plasma glucose levels and the overall glycemic excursion (area under the curve) to a glucose load were significantly reduced (1.6-fold; P < 0.05) in (Pro3)GIP-treated mice compared with controls. GIP-R ablation also significantly lowered overall plasma glucose (1.4-fold; P < 0.05) and insulin (1.5-fold; P < 0.05) responses to feeding. These changes were associated with significantly enhanced (1.6-fold; P < 0.05) insulin sensitivity in the (Pro3)GIP-treated group. Daily injection of (Pro3)GIP reduced pancreatic insulin content (1.3-fold; P < 0.05) and partially corrected the obesity-related islet hypertrophy and beta-cell hyperplasia of ob/ob mice. These comprehensive beneficial effects of (Pro3)GIP were reversed 9 days after cessation of treatment and were independent of food intake and body weight, which were unchanged. These studies highlight a role for GIP in obesity-related glucose intolerance and emphasize the potential of specific GIP-R antagonists as a new class of drugs for the alleviation of insulin resistance and treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/administration & dosage , Insulin Resistance , Islets of Langerhans/pathology , Obesity/complications , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Blood Glucose/analysis , Body Weight/drug effects , Diabetes Mellitus, Type 2/etiology , Eating/drug effects , Food , Glucose Intolerance/drug therapy , Glycated Hemoglobin/analysis , Hyperplasia , Insulin/analysis , Insulin/blood , Islets of Langerhans/chemistry , Kinetics , Mice , Mice, Obese , Receptors, Gastrointestinal Hormone/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...