Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Mol Metab ; 88: 102006, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39128651

ABSTRACT

OBJECTIVES: Obesity represents a global health crisis with significant patient burdens and healthcare costs. Despite the advances with glucagon-like peptide-1 (GLP-1) receptor agonists in treating obesity, unmet needs remain. This study characterizes a novel glucose-dependent insulinotropic polypeptide receptor (GIPR) peptide antagonist, AT-7687, evaluating its potential to enhance obesity treatment. METHODS: We assessed the in vitro potency and pharmacokinetics of AT-7687, alongside its therapeutic effects when administered subcutaneously (SC) alone and in combination with liraglutide to high-fat-diet-fed obese non-human primates (NHP). The study spanned a 42-day treatment period and a 15-day washout period. RESULTS: AT-7687 demonstrated a subnanomolar cAMP antagonistic potency (pKB of 9.5) in HEK-293 cells and a 27.4 h half-life in NHPs. It effectively maintained weight stability in obese monkeys, whereas placebo recipients had an 8.6% weight increase by day 42 (P = 0.01). Monotherapy with liraglutide resulted in a 12.4% weight reduction compared to placebo (P = 0.03) and combining AT-7687 with liraglutide led to a 16.3% weight reduction (P = 0.0002). The combination therapy significantly improved metabolic markers, reducing insulin levels by 52% (P = 0.008), glucose by 30% (P = 0.02), triglycerides by 39% (P = 0.05), total cholesterol by 29% (P = 0.03), and LDL cholesterol by 48% (P = 0.003) compared to placebo. AT-7687 treatment was well tolerated and not associated with any side effects. CONCLUSIONS: This study underscores the potential of AT-7687 as a promising addition to current obesity treatments.


Subject(s)
Liraglutide , Macaca fascicularis , Obesity , Receptors, Gastrointestinal Hormone , Weight Loss , Animals , Humans , Liraglutide/pharmacology , Liraglutide/administration & dosage , Weight Loss/drug effects , Obesity/drug therapy , Obesity/metabolism , Male , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , HEK293 Cells , Diet, High-Fat/adverse effects , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide 1 , Blood Glucose/drug effects , Blood Glucose/metabolism
2.
J Endocrinol ; 262(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38861364

ABSTRACT

Recent approval of the dual glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) receptor agonist, tirzepatide, for the management of type 2 diabetes mellitus (T2DM) has reinvigorated interest in exploitation of GIP receptor (GIPR) pathways as a means of metabolic disease management. However, debate has long surrounded the use of the GIPR as a therapeutic target and whether agonism or antagonism is of most benefit in management of obesity/diabetes. This controversy appears to be partly resolved by the success of tirzepatide. However, emerging studies indicate that prolonged GIPR agonism may desensitise the GIPR to essentially induce receptor antagonism, with this phenomenon suggested to be more pronounced in the human than rodent setting. Thus, deliberation continues to rage in relation to benefits of GIPR agonism vs antagonism. That said, as with GIPR agonism, it is clear that the metabolic advantages of sustained GIPR antagonism in obesity and obesity-driven forms of diabetes can be enhanced by concurrent GLP-1 receptor (GLP-1R) activation. This narrative review discusses various approaches of pharmacological GIPR antagonism including small molecule, peptide, monoclonal antibody and peptide-antibody conjugates, indicating stage of development and significance to the field. Taken together, there is little doubt that interesting times lie ahead for GIPR agonism and antagonism, either alone or when combined with GLP-1R agonists, as a therapeutic intervention for the management of obesity and associated metabolic disease.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Obesity , Receptors, Gastrointestinal Hormone , Humans , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism , Obesity/drug therapy , Obesity/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Gastric Inhibitory Polypeptide/agonists , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Glucagon-Like Peptide-2 Receptor
3.
Trends Endocrinol Metab ; 35(7): 566-568, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38763780

ABSTRACT

Unimolecular co-agonists at the GLP-1/GIP receptors have recently achieved remarkable anti-obesogenic feats; yet, in a recent Phase 1 clinical trial, Véniant and colleagues report astounding body-weight loss, and an appreciable safety profile, in participants with obesity using the GLP-1R agonist/GIPR antagonist AMG 133.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Obesity , Receptors, Gastrointestinal Hormone , Humans , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism , Obesity/metabolism , Peptides/pharmacology , Animals , Weight Loss/drug effects , Receptors, Glucagon/metabolism , Receptors, Glucagon/antagonists & inhibitors
4.
Peptides ; 177: 171212, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38608836

ABSTRACT

Surprisingly, agonists, as well as antagonists of the glucose-dependent insulinotropic polypeptide receptor (GIPR), are currently being used or investigated as treatment options for type 2 diabetes and obesity - and both, when combined with glucagon-like peptide 1 receptor (GLP-1R) agonism, enhance GLP-1-induced glycemia and weight loss further. This paradox raises several questions regarding not only the mechanisms of actions of GIP but also the processes engaged during the activation of both the GIP and GLP-1 receptors. Here, we provide an overview of studies of the properties and actions of peptide-derived GIPR antagonists, focusing on GIP(3-30)NH2, a naturally occurring N- and C-terminal truncation of GIP(1-42). GIP(3-30)NH2 was the first GIPR antagonist administered to humans. GIP(3-30)NH2 and a few additional antagonists, like Pro3-GIP, have been used in both in vitro and in vivo studies to elucidate the molecular and cellular consequences of GIPR inhibition, desensitization, and internalization and, at a larger scale, the role of the GIP system in health and disease. We provide an overview of these studies combined with recent knowledge regarding the effects of naturally occurring variants of the GIPR system and species differences within the GIP system to enhance our understanding of the GIPR as a drug target.


Subject(s)
Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism , Humans , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/chemistry , Animals , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Obesity/drug therapy , Obesity/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Peptide Fragments/pharmacology , Peptide Fragments/chemistry , Peptide Fragments/metabolism
5.
Mol Metab ; 66: 101638, 2022 12.
Article in English | MEDLINE | ID: mdl-36400403

ABSTRACT

OBJECTIVE: Glucose-dependent insulinotropic polypeptide (GIP) is one of the two major incretin factors that regulate metabolic homeostasis. Genetic ablation of its receptor (GIPR) in mice confers protection against diet-induced obesity (DIO), while GIPR neutralizing antibodies produce additive weight reduction when combined with GLP-1R agonists in preclinical models and clinical trials. Conversely, GIPR agonists have been shown to promote weight loss in rodents, while dual GLP-1R/GIPR agonists have proven superior to GLP-1R monoagonists for weight reduction in clinical trials. We sought to develop a long-acting, specific GIPR peptide antagonist as a tool compound suitable for investigating GIPR pharmacology in both rodent and human systems. METHODS: We report a structure-activity relationship of GIPR peptide antagonists based on the human and mouse GIP sequences with fatty acid-based protraction. We assessed these compounds in vitro, in vivo in DIO mice, and ex vivo in islets from human donors. RESULTS: We report the discovery of a GIP(5-31) palmitoylated analogue, [Nα-Ac, L14, R18, E21] hGIP(5-31)-K11 (γE-C16), which potently inhibits in vitro GIP-mediated cAMP generation at both the hGIPR and mGIPR. In vivo, this peptide effectively blocks GIP-mediated reductions in glycemia in response to exogenous and endogenous GIP and displays a circulating pharmacokinetic profile amenable for once-daily dosing in rodents. Co-administration with the GLP-1R agonist semaglutide and this GIPR peptide antagonist potentiates weight loss compared to semaglutide alone. Finally, this antagonist inhibits GIP- but not GLP-1-stimulated insulin secretion in intact human islets. CONCLUSIONS: Our work demonstrates the discovery of a potent, specific, and long-acting GIPR peptide antagonist that effectively blocks GIP action in vitro, ex vivo in human islets, and in vivo in mice while producing additive weight-loss when combined with a GLP-1R agonist in DIO mice.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Receptors, Gastrointestinal Hormone , Rodentia , Animals , Humans , Mice , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Mice, Obese , Peptides/pharmacology , Peptides/chemistry , Rodentia/metabolism , Weight Loss , Receptors, Gastrointestinal Hormone/antagonists & inhibitors
6.
Front Endocrinol (Lausanne) ; 13: 887238, 2022.
Article in English | MEDLINE | ID: mdl-35712239

ABSTRACT

Background: Chronic pain is defined as pain that persists typically for a period of over six months. Chronic pain is often accompanied by an anxiety disorder, and these two tend to exacerbate each other. This can make the treatment of these conditions more difficult. Glucose-dependent insulinotropic polypeptide (GIP) is a member of the incretin hormone family and plays a critical role in glucose metabolism. Previous research has demonstrated the multiple roles of GIP in both physiological and pathological processes. In the central nervous system (CNS), studies of GIP are mainly focused on neurodegenerative diseases; hence, little is known about the functions of GIP in chronic pain and pain-related anxiety disorders. Methods: The chronic inflammatory pain model was established by hind paw injection with complete Freund's adjuvant (CFA) in C57BL/6 mice. GIP receptor (GIPR) agonist (D-Ala2-GIP) and antagonist (Pro3-GIP) were given by intraperitoneal injection or anterior cingulate cortex (ACC) local microinjection. Von Frey filaments and radiant heat were employed to assess the mechanical and thermal hypersensitivity. Anxiety-like behaviors were detected by open field and elevated plus maze tests. The underlying mechanisms in the peripheral nervous system and CNS were explored by GIPR shRNA knockdown in the ACC, enzyme-linked immunosorbent assay, western blot analysis, whole-cell patch-clamp recording, immunofluorescence staining and quantitative real-time PCR. Results: In the present study, we found that hind paw injection with CFA induced pain sensitization and anxiety-like behaviors in mice. The expression of GIPR in the ACC was significantly higher in CFA-injected mice. D-Ala2-GIP administration by intraperitoneal or ACC local microinjection produced analgesic and anxiolytic effects; these were blocked by Pro3-GIP and GIPR shRNA knockdown in the ACC. Activation of GIPR inhibited neuroinflammation and activation of microglia, reversed the upregulation of NMDA and AMPA receptors, and suppressed the enhancement of excitatory neurotransmission in the ACC of model mice. Conclusions: GIPR activation was found to produce analgesic and anxiolytic effects, which were partially due to attenuation of neuroinflammation and inhibition of excitatory transmission in the ACC. GIPR may be a suitable target for treatment of chronic inflammatory pain and pain-related anxiety.


Subject(s)
Chronic Pain , Receptors, Gastrointestinal Hormone , Animals , Chronic Pain/drug therapy , Chronic Pain/metabolism , Freund's Adjuvant , Gastric Inhibitory Polypeptide/physiology , Gyrus Cinguli/metabolism , Mice , Mice, Inbred C57BL , RNA, Small Interfering , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/metabolism
7.
Toxins (Basel) ; 13(8)2021 07 22.
Article in English | MEDLINE | ID: mdl-34437383

ABSTRACT

Deoxynivalenol (DON), also known as vomitoxin, is a mycotoxin that can cause antifeeding and vomiting in animals. However, the mechanism of DON inducing anorexia is complicated. Studies have shown that intestinal hormones play a significant part in the anorexia caused by DON. We adopted the "modeling of acute antifeeding in mice" as the basic experimental model, and used two methods of gavage and intraperitoneal injection to explore the effect of intestinal hormones on the antifeedant response induced by DON in mice. We found that 1 and 2.5 mg/kg·bw of DON can acutely induce anorexia and increase the plasma intestinal hormones CCK, PYY, GIP, and GLP-1 in mice within 3 h. Direct injection of exogenous intestinal hormones CCK, PYY, GIP, and GLP-1 can trigger anorexia behavior in mice. Furthermore, the PYY receptor antagonist JNJ-31020028, GLP-1 receptor antagonist Exendin(9-39), CCK receptor antagonist Proglumide, GIP receptor antagonist GIP(3-30)NH2 attenuated both intestinal hormone and DON-induced anorectic responses. These results indicate that intestinal hormones play a critical role in the anorexia response induced by DON.


Subject(s)
Anorexia/chemically induced , Gastrointestinal Hormones/blood , Trichothecenes/toxicity , Animals , Anorexia/drug therapy , Anorexia/metabolism , Benzamides/therapeutic use , Eating/drug effects , Feeding Behavior/drug effects , Female , Gastric Inhibitory Polypeptide/therapeutic use , Mice , Peptide Fragments/therapeutic use , Piperazines/therapeutic use , Proglumide/therapeutic use , Receptors, Gastrointestinal Hormone/antagonists & inhibitors
8.
Cell Rep Med ; 2(5): 100263, 2021 05 18.
Article in English | MEDLINE | ID: mdl-34095876

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) regulate glucose and energy homeostasis. Targeting both pathways with GIP receptor (GIPR) antagonist antibody (GIPR-Ab) and GLP-1 receptor (GLP-1R) agonist, by generating GIPR-Ab/GLP-1 bispecific molecules, is an approach for treating obesity and its comorbidities. In mice and monkeys, these molecules reduce body weight (BW) and improve many metabolic parameters. BW loss is greater with GIPR-Ab/GLP-1 than with GIPR-Ab or a control antibody conjugate, suggesting synergistic effects. GIPR-Ab/GLP-1 also reduces the respiratory exchange ratio in DIO mice. Simultaneous receptor binding and rapid receptor internalization by GIPR-Ab/GLP-1 amplify endosomal cAMP production in recombinant cells expressing both receptors. This may explain the efficacy of the bispecific molecules. Overall, our GIPR-Ab/GLP-1 molecules promote BW loss, and they may be used for treating obesity.


Subject(s)
Body Weight/physiology , Glucagon-Like Peptide 1/metabolism , Obesity/metabolism , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Tolerance Test/methods , Haplorhini/metabolism , Mice, Obese
9.
Eur J Endocrinol ; 185(1): 33-45, 2021 May 21.
Article in English | MEDLINE | ID: mdl-33886495

ABSTRACT

OBJECTIVE: The insulinotropic effect of exogenous, intravenously infused glucose-dependent insulinotropic polypeptide (GIP) is impaired in patients with type 2 diabetes. We evaluated the effects of endogenous GIP in relation to glucose and bone metabolism in patients with type 2 diabetes using a selective GIP receptor antagonist and hypothesized that the effects of endogenous GIP were preserved. DESIGN: A randomized, double-blinded, placebo-controlled, crossover study. METHODS: Ten patients with overweight/obesity and type 2 diabetes (mean±s.d.; HbA1c 52 ± 11 mmol/mol; BMI 32.5 ± 4.8 kg/m2) were included. We infused a selective GIP receptor antagonist, GIP(3-30)NH2 (1200 pmol/kg/min), or placebo (saline) during two separate, 230-min, standardized, liquid mixed meal tests followed by a meal ad libitum. Subcutaneous adipose tissue biopsies were analyzed. RESULTS: Compared with placebo, GIP(3-30)NH2 reduced postprandial insulin secretion (Δbaseline-subtracted area under the curve (bsAUC)C-peptide% ± s.e.m.; -14 ± 6%, P = 0.021) and peak glucagon (Δ% ± s.e.m.; -11 ± 6%, P = 0.046) but had no effect on plasma glucose (P = 0.692). Suppression of bone resorption (assessed by circulating carboxy-terminal collagen crosslinks (CTX)) was impaired during GIP(3-30)NH2 infusion compared with placebo (ΔbsAUCCTX; ±s.e.m.; -4.9 ± 2 ng/mL × min, P = 0.005) corresponding to a ~50% reduction. Compared with placebo, GIP(3-30)NH2 did not affect plasma lipids, meal consumption ad libitum or adipose tissue triglyceride content. CONCLUSIONS: Using a selective GIP receptor antagonist during a meal, we show that endogenous GIP increases postprandial insulin secretion with little effect on postprandial glycaemia but is important for postprandial bone homeostasis in patients with type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Bone Resorption/metabolism , Diabetes Mellitus, Type 2/metabolism , Gastric Inhibitory Polypeptide/metabolism , Insulin Secretion/physiology , Obesity/metabolism , Triglycerides/metabolism , Adult , Aged , Blood Glucose/drug effects , Collagen Type I/drug effects , Collagen Type I/metabolism , Cross-Over Studies , Double-Blind Method , Feeding Behavior/drug effects , Gastric Inhibitory Polypeptide/pharmacology , Humans , Insulin Secretion/drug effects , Male , Middle Aged , Peptide Fragments/pharmacology , Peptides/drug effects , Peptides/metabolism , Postprandial Period , Random Allocation , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism
10.
Structure ; 29(7): 679-693.e6, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33891864

ABSTRACT

The glucose-dependent insulinotropic polypeptide (GIP) is a 42-residue metabolic hormone that is actively being targeted for its regulatory role of glycemia and energy balance. Limited structural data of its receptor has made ligand design tedious. This study investigates the structure and function of the GIP receptor (GIPR), using a homology model based on the GLP-1 receptor. Molecular dynamics combined with in vitro mutational data were used to pinpoint residues involved in ligand binding and/or receptor activation. Significant differences in binding mode were identified for the naturally occurring agonists GIP(1-30)NH2 and GIP(1-42) compared with high potency antagonists GIP(3-30)NH2 and GIP(5-30)NH2. Residues R1832.60, R1902.67, and R3005.40 are shown to be key for activation of the GIPR, and evidence suggests that a disruption of the K293ECL2-E362ECL3 salt bridge by GIPR antagonists strongly reduces GIPR activation. Combinatorial use of these findings can benefit rational design of ligands targeting the GIPR.


Subject(s)
Glucagon-Like Peptide-1 Receptor/chemistry , Glucagon-Like Peptide-1 Receptor/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Binding Sites , Gastric Inhibitory Polypeptide/metabolism , Humans , Hydrogen Bonding , Models, Molecular , Molecular Dynamics Simulation , Mutation , Protein Conformation , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/genetics , Structural Homology, Protein
11.
Endocrinology ; 162(7)2021 07 01.
Article in English | MEDLINE | ID: mdl-33782700

ABSTRACT

The incretin effect-the amplification of insulin secretion after oral vs intravenous administration of glucose as a mean to improve glucose tolerance-was suspected even before insulin was discovered, and today we know that the effect is due to the secretion of 2 insulinotropic peptides, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1). But how important is it? Physiological experiments have shown that, because of the incretin effect, we can ingest increasing amounts of amounts of glucose (carbohydrates) without increasing postprandial glucose excursions, which otherwise might have severe consequences. The mechanism behind this is incretin-stimulated insulin secretion. The availability of antagonists for GLP-1 and most recently also for GIP has made it possible to directly estimate the individual contributions to postprandial insulin secretion of a) glucose itself: 26%; b) GIP: 45%; and c) GLP-1: 29%. Thus, in healthy individuals, GIP is the champion. When the action of both incretins is prevented, glucose tolerance is pathologically impaired. Thus, after 100 years of research, we now know that insulinotropic hormones from the gut are indispensable for normal glucose tolerance. The loss of the incretin effect in type 2 diabetes, therefore, contributes greatly to the impaired postprandial glucose control.


Subject(s)
Blood Glucose/physiology , Gastric Inhibitory Polypeptide/physiology , Glucagon-Like Peptide 1/physiology , Homeostasis/physiology , Incretins/physiology , Insulin/physiology , Diabetes Mellitus, Type 2/physiopathology , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Glucagon-Like Peptide 1/antagonists & inhibitors , Humans , Insulin Secretion/drug effects , Postprandial Period , Receptors, Gastrointestinal Hormone/antagonists & inhibitors
12.
Life Sci ; 269: 119038, 2021 Mar 15.
Article in English | MEDLINE | ID: mdl-33453239

ABSTRACT

OBJECTIVE: Glucose-dependent insulinotropic polypeptide receptor (GIPR) has been identified as a contributor to obesity, and GIPR knockout mice are protected against diet-induced obesity (DIO). Therefore, we developed the anti-GIPR antagonistic monoclonal antibody (mAb) alone and in combination with DPP-4 inhibitor as potential therapeutic strategy for treating obesity and dyslipidemia based on this genetic evidence. METHODS: Fully neutralized GIPR activity of GIPR-monoclonal antibody (mAb) was assessed by regulating the in vitro production of cAMP in the mouse GIPR stably expressing cells. Chronic efficacies of GIPR-mAb alone and in combination with DPP-4 inhibitor Sitagliptin in diabetic or DIO mice were both investigated. Multiple metabolic parameters including body weight, glucose level, fat mass, lipid metabolism-related indicators as well as H&E staining and immunohistochemical analysis were performed. Role of GIPR in pancreatic cells on regulating fat metabolism was explored in GIPR ß-cell knockout mouse model. RESULTS: Chronic treatment of GIPR-mAb improved body weight control, glucose metabolism, and was associated with reduced fat mass, enhanced pancreatic function and exchange ratio of the resting respiratory in diabetic mice. In addition, further study of anti-GIPR mAb combined with Sitagliptin in DIO mice demonstrated significantly improved weight loss compare to the both monomer treatment. Furthermore, we demonstrated important role of GIPR in ß-cell in regulating the fat mass and response to antagonistic GIPR-mAb in a conditional GIPR-knockout mouse. CONCLUSION: Chronic treatment with anti-GIPR mAb alone and combined with DPP-4 inhibitor provide preclinical therapeutic approaches to treat obesity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Dipeptidyl Peptidase 4/chemistry , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dyslipidemias/drug therapy , Kidney Diseases/drug therapy , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Animals , Body Weight , Diet, High-Fat/adverse effects , Drug Therapy, Combination , Dyslipidemias/etiology , Dyslipidemias/pathology , Kidney Diseases/etiology , Kidney Diseases/pathology , Male , Mice , Mice, Knockout , Mice, Obese , Obesity/etiology , Obesity/pathology , Receptors, Gastrointestinal Hormone/immunology , Receptors, Gastrointestinal Hormone/physiology , Weight Loss
13.
SLAS Discov ; 26(1): 1-16, 2021 01.
Article in English | MEDLINE | ID: mdl-32749201

ABSTRACT

The secretin receptor (SCTR), a prototypical class B G protein-coupled receptor (GPCR), exerts its effects mainly by activating Gαs proteins upon binding of its endogenous peptide ligand secretin. SCTRs can be found in a variety of tissues and organs across species, including the pancreas, stomach, liver, heart, lung, colon, kidney, and brain. Beyond that, modulation of SCTR-mediated signaling has therapeutic potential for the treatment of multiple diseases, such as heart failure, obesity, and diabetes. However, no ligands other than secretin and its peptide analogs have been described to regulate SCTRs, probably due to inherent challenges in family B GPCR drug discovery. Here we report creation of a testing funnel that allowed targeted detection of SCTR small-molecule activators. Pursuing the strategy to identify positive allosteric modulators (PAMs), we established a unique primary screening assay employing a mixture of three orthosteric stimulators that was compared in a screening campaign testing 12,000 small-molecule compounds. Beyond that, we developed a comprehensive set of secondary assays, such as a radiolabel-free target engagement assay and a NanoBiT (NanoLuc Binary Technology)-based approach to detect ß-arrestin-2 recruitment, all feasible in a high-throughput environment as well as capable of profiling ligands and hits regarding their effect on binding and receptor function. This combination of methods enabled the discovery of five promising scaffolds, four of which have been validated and further characterized with respect to their allosteric activities. We propose that our results may serve as starting points for developing the first in vivo active small molecules targeting SCTRs.


Subject(s)
Drug Development/methods , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/chemistry , Animals , Biobehavioral Sciences , CHO Cells , Calcium/metabolism , Carrier Proteins , Cricetulus , Cyclic AMP/metabolism , Gene Expression , Genes, Reporter , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Ligands , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Conformation , Structure-Activity Relationship
14.
Nat Commun ; 11(1): 4981, 2020 10 05.
Article in English | MEDLINE | ID: mdl-33020469

ABSTRACT

Antagonism or agonism of the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) prevents weight gain and leads to dramatic weight loss in combination with glucagon-like peptide-1 receptor agonists in preclinical models. Based on the genetic evidence supporting GIPR antagonism, we previously developed a mouse anti-murine GIPR antibody (muGIPR-Ab) that protected diet-induced obese (DIO) mice against body weight gain and improved multiple metabolic parameters. This work reconciles the similar preclinical body weight effects of GIPR antagonists and agonists in vivo, and here we show that chronic GIPR agonism desensitizes GIPR activity in primary adipocytes, both differentiated in vitro and adipose tissue in vivo, and functions like a GIPR antagonist. Additionally, GIPR activity in adipocytes is partially responsible for muGIPR-Ab to prevent weight gain in DIO mice, demonstrating a role of adipocyte GIPR in the regulation of adiposity in vivo.


Subject(s)
Adipocytes/drug effects , Anti-Obesity Agents/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Adipocytes/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/therapeutic use , Antibodies/pharmacology , Antibodies/therapeutic use , Body Weight/drug effects , Cell Membrane/metabolism , Cells, Cultured , Cyclic AMP/metabolism , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/metabolism
15.
Adv Clin Exp Med ; 29(6): 661-668, 2020 06.
Article in English | MEDLINE | ID: mdl-32614526

ABSTRACT

BACKGROUND: New glucagon-like peptide-1 (GLP-1) analogues developed in recent years have a long half-life and offer further prospects for clinical application. At present, the neuroprotection of GLP-1 analogues in Alzheimer's disease (AD) has just begun to be explored. OBJECTIVES: To investigate how glucagon-like peptide-1 (liraglutide) plays a protective role in AD by regulating tau activation and BACE1 expression. MATERIAL AND METHODS: Human neuroblastoma cell line SH-SY5Y cells were cultured in vitro and pretreated with different concentrations of liraglutide, and then treated with different concentrations of okadaic acid (OA) in order to observe the apoptosis of the SH-SY5Y cells. After liraglutide treatment, the apoptosis of neurons in AD rats was detected using flow cytometry, and tau activation and ß-site APP cleaving enzyme 1 (BACE1) expression were detected using western blot. RESULTS: Different concentrations of OA were able to induce apoptosis of SH-SY5Y cells in a dose-dependent manner. Different concentrations of liraglutide were used to pretreat SH-SY5Y cells, which were able to protect the SH-SY5Y cells from apoptosis induced by OA. Okadaic acid significantly increased tau activation and BACE1 expression in the SH-SY5Y cells, which was blocked with liraglutide pretreatment. The results of a water maze experiment showed that liraglutide had significant protective effects on memory and cognitive ability in AD rats induced with OA, inhibited apoptosis of neural cells in AD rats, and inhibited tau activation and BACE1 expression of neural cells in AD rats induced with OA. CONCLUSIONS: Liraglutide has a protective effect on AD in vivo and in vitro, which may be mediated by preventing neuronal apoptosis and inhibiting the activation of tau and the expression of BACE1.


Subject(s)
Alzheimer Disease , Liraglutide , Neuroprotective Agents , Receptors, Gastrointestinal Hormone , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/drug effects , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/therapeutic use , Animals , Aspartic Acid Endopeptidases/drug effects , Aspartic Acid Endopeptidases/metabolism , Aspartic Acid Endopeptidases/therapeutic use , Cell Line, Tumor , Glucagon-Like Peptide 1/therapeutic use , Humans , Liraglutide/pharmacology , Neuroprotective Agents/pharmacology , Rats , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors
16.
J Clin Endocrinol Metab ; 105(8)2020 08 01.
Article in English | MEDLINE | ID: mdl-32459834

ABSTRACT

The 2 hormones responsible for the amplification of insulin secretion after oral as opposed to intravenous nutrient administration are the gut peptides, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP). However, whereas GLP-1 also inhibits appetite and food intake and improves glucose regulation in patients with type 2 diabetes (T2DM), GIP seems to be devoid of these activities, although the 2 hormones as well as their receptors are highly related. In fact, numerous studies have suggested that GIP may promote obesity. However, chimeric peptides, combining elements of both peptides and capable of activating both receptors, have recently been demonstrated to have remarkable weight-losing and glucose-lowering efficacy in obese individuals with T2DM. At the same time, antagonists of the GIP receptor have been reported to reduce weight gain/cause weight loss in experimental animals including nonhuman primates. This suggests that both agonists and antagonist of the GIP receptor should be useful, at least for weight-losing therapy. How is this possible? We here review recent experimental evidence that agonist-induced internalization of the two receptors differs markedly and that modifications of the ligand structures, as in co-agonists, profoundly influence these cellular processes and may explain that an antagonist may activate while an agonist may block receptor signaling.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/metabolism , Obesity/drug therapy , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Appetite/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/metabolism , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Incretins/agonists , Incretins/metabolism , Obesity/blood , Obesity/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Signal Transduction/drug effects , Weight Loss/drug effects
17.
Biochem Pharmacol ; 177: 113929, 2020 07.
Article in English | MEDLINE | ID: mdl-32217097

ABSTRACT

The secretin receptor is a prototypic class B GPCR with substantial and broad pharmacologic importance. The aim of this project was to develop a high affinity selective antagonist as a new and important pharmacologic tool and to aid stabilization of this receptor in an inactive conformation for ultimate structural characterization. Amino-terminal truncation of the natural 27-residue ligand reduced biological activity, but also markedly reduced binding affinity. This was rationally and experimentally overcome with lactam stabilization of helical structure and with replacement of residues with natural and unnatural amino acids. A key new step in this effort was the replacement of peptide residue Leu22 with L-cyclohexylalanine (Cha) to enhance potential hydrophobic interactions with receptor residues Leu31, Val34, and Phe92 that were predicted from molecular modeling. Alanine-replacement mutagenesis of these residues markedly affected ligand binding and biological activity. The optimal antagonist ligand, (Y10,c[E16,K20],I17,Cha22,R25)sec(6-27), exhibited high binding affinity (4 nM), similar to natural secretin, and exhibited no demonstrable biological activity to stimulate cAMP accumulation, intracellular calcium mobilization, or ß-arrestin-2 translocation. It acts as an orthosteric competitive antagonist, predicted to bind within the peptide-binding groove in the receptor extracellular domain. The analogous peptide that was one residue longer, retaining Thr5, exhibited partial agonist activity, while further truncation of even a single residue (Phe6) reduced binding affinity. This sec(6-27)-based peptide will be an important new tool for pharmacological and structural studies.


Subject(s)
Drug Design , Peptides/chemistry , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/chemistry , Secretin/analogs & derivatives , Alanine/metabolism , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cricetulus , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Models, Molecular , Peptides/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Receptors, Calcitonin/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Secretin/metabolism
18.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Article in English | MEDLINE | ID: mdl-32077470

ABSTRACT

CONTEXT: The actions of both endogenous incretin hormones during a meal have not previously been characterized. OBJECTIVE: Using specific receptor antagonists, we investigated the individual and combined contributions of endogenous glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) to postprandial glucose metabolism, energy expenditure, and gallbladder motility. DESIGN: Randomized, double-blinded, placebo-controlled, crossover design. SETTING: On four separate days, four liquid mixed meal tests (1894 kJ) over 270 minutes (min). PATIENTS OR OTHER PARTICIPANTS: Twelve healthy male volunteers. INTERVENTIONS: Infusions of the GIP receptor antagonist GIP(3-30)NH2 (800 pmol/kg/min), the GLP-1 receptor antagonist exendin(9-39)NH2 (0-20 min: 1000 pmol/kg/min; 20-270 min: 450 pmol/kg/min), GIP(3-30)NH2+exendin(9-39)NH2, or placebo/saline. MAIN OUTCOME MEASURE: Baseline-subtracted area under the curve (bsAUC) of C-peptide. RESULTS: Infusion of GIP(3-30)NH2+exendin(9-39)NH2 significantly increased plasma glucose excursions (bsAUC: 261 ± 142 mmol/L × min) during the liquid mixed meals compared with GIP(3-30)NH2 (180 ± 141 mmol/L × min; P = 0.048), exendin(9-39)NH2 (171 ± 114 mmol/L × min; P = 0.046), and placebo (116 ± 154 mmol/L × min; P = 0.015). Correspondingly, C-peptide:glucose ratios during GIP(3-30)NH2+exendin(9-39)NH2 infusion were significantly lower than during GIP(3-30)NH2 (P = 0.0057), exendin(9-39)NH2 (P = 0.0038), and placebo infusion (P = 0.014). GIP(3-30)NH2 resulted in significantly lower AUCs for glucagon than exendin(9-39)NH2 (P = 0.0417). Gallbladder ejection fraction was higher during GIP(3-30)NH2 compared with placebo (P = 0.004). For all interventions, energy expenditure and respiratory quotient were similar. CONCLUSIONS: Endogenous GIP and GLP-1 lower postprandial plasma glucose excursions and stimulate insulin secretion but only endogenous GIP affects gallbladder motility. The two incretin hormones potentiate each other's effects in the control of postprandial glycemia in healthy men.


Subject(s)
Blood Glucose/drug effects , Energy Metabolism/drug effects , Gastrointestinal Motility/drug effects , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Adult , Aged , Cross-Over Studies , Double-Blind Method , Gallbladder/metabolism , Gastric Inhibitory Polypeptide/administration & dosage , Healthy Volunteers , Humans , Incretins/blood , Male , Meals , Middle Aged , Peptide Fragments/administration & dosage , Postprandial Period/drug effects , Young Adult
19.
MAbs ; 12(1): 1710047, 2020.
Article in English | MEDLINE | ID: mdl-31905038

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone involved in regulating glucose and lipid metabolism. GIP receptor (GIPR) antagonism is believed to offer therapeutic potential for various metabolic diseases. Pharmacological intervention of GIPR, however, has limited success due to lack of effective antagonistic reagents. Previously we reported the discovery of two mouse anti-murine GIPR monoclonal antibodies (mAbs) with distinctive properties in rodent models. Here, we report the detailed structural and biochemical characterization of these two antibodies, mAb1 and mAb2. In vitro and in vivo characterizations demonstrated mAb2 is a full GIPR antagonistic antibody and mAb1 is a non-neutralizing GIPR binder. To understand the molecular basis of these two antibodies, we determined the co-crystal structures of GIPR extracellular domain in complex with mAb1 and with mAb2 at resolutions of 2.1 and 2.6 Å, respectively. While the non-neutralizing mAb1 binds to GIPR without competing with the ligand peptide, mAb2 not only partially occludes the ligand peptide binding, but also recognizes the GIPR C-terminal stalk region in a helical conformation that acts as a molecular mimic of the ligand peptide and locks GIPR in a novel auto-inhibited state. Furthermore, administration of mAb2 in diet-induced obesity mice for 7 weeks leads to both reduction in body weight gain and improvement of metabolic profiles. In contrast, mAb1 has no effect on body weight or other metabolic improvement. Together, our studies reveal the unique molecular mechanism of action underlying the superior antagonistic activity of mAb2 and signify the promising therapeutic potential of effective GIPR antagonism for the treatment of metabolic disorders.


Subject(s)
Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/pharmacology , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Weight Gain/drug effects , Animals , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Protein Conformation
20.
Peptides ; 125: 170228, 2020 03.
Article in English | MEDLINE | ID: mdl-31812593

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a gut hormone secreted primarily from enteroendocrine K cells in the duodenum and proximal jejunum following nutrient ingestion, primarily acting on islet ß-cells to potentiate insulin secretion in a glucose-dependent manner. New discoveries of GIP receptor (GIPR) biology in adipose tissue, as well as findings that co-agonists for the glucagon-like peptide-1 receptor (GLP-1R) and GIPR induce greater weight loss than that seen with GLP-1R agonists alone, has led to continued interest in manipulating GIPR activity for the treatment of obesity/type 2 diabetes mellitus (T2DM). As cardiovascular diseases represent the number one cause of death in people with T2DM, there has also been growing interest in understanding the cardiovascular actions of the GIP/GIPR axis. Although controversy surrounds whether GIPR agonism or antagonism will induce weight loss in obesity/T2DM, such actions undoubtedly will impact cardiovascular function and outcomes since obesity is a major risk factor for cardiovascular disease. Furthermore, GIPR agonism is associated with reduced progression of atherosclerotic lesions in preclinical studies. Conversely, genetic deletion of GIPR activity within cardiac myocytes of the heart results in robust protection against experimental myocardial infarction. Nonetheless, interrogation of the GIP/GIPR axis on cardiac function in humans will involve the systemic actions of the GIPR within the myocardium and other systems (e.g. adipose tissue, vasculature), which will influence the long-term future of GIPR modification for the treatment of obesity/T2DM.


Subject(s)
Cardiovascular System/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Gastrointestinal Agents/pharmacology , Myocytes, Cardiac/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Animals , Cardiovascular System/drug effects , Humans , Insulin Secretion , Myocytes, Cardiac/drug effects , Receptors, Gastrointestinal Hormone/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL