Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 120(41): e2306145120, 2023 10 10.
Article in English | MEDLINE | ID: mdl-37792509

ABSTRACT

Glucose-dependent insulinotropic polypeptide receptor (GIPR) is a potential drug target for metabolic disorders. It works with glucagon-like peptide-1 receptor and glucagon receptor in humans to maintain glucose homeostasis. Unlike the other two receptors, GIPR has at least 13 reported splice variants (SVs), more than half of which have sequence variations at either C or N terminus. To explore their roles in endogenous peptide-mediated GIPR signaling, we determined the cryoelectron microscopy (cryo-EM) structures of the two N terminus-altered SVs (referred as GIPR-202 and GIPR-209 in the Ensembl database, SV1 and SV2 here, respectively) and investigated the outcome of coexpressing each of them in question with GIPR in HEK293T cells with respect to ligand binding, receptor expression, cAMP (adenosine 3,5-cyclic monophosphate) accumulation, ß-arrestin recruitment, and cell surface localization. It was found that while both N terminus-altered SVs of GIPR neither bound to the hormone nor elicited signal transduction per se, they suppressed ligand binding and cAMP accumulation of GIPR. Meanwhile, SV1 reduced GIPR-mediated ß-arrestin 2 responses. The cryo-EM structures of SV1 and SV2 showed that they reorganized the extracellular halves of transmembrane helices 1, 6, and 7 and extracellular loops 2 and 3 to adopt a ligand-binding pocket-occupied conformation, thereby losing binding ability to the peptide. The results suggest a form of signal bias that is constitutive and ligand-independent, thus expanding our knowledge of biased signaling beyond pharmacological manipulation (i.e., ligand specific) as well as constitutive and ligand-independent (e.g., SV1 of the growth hormone-releasing hormone receptor).


Subject(s)
Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Humans , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Gastric Inhibitory Polypeptide/pharmacology , Ligands , Cryoelectron Microscopy , HEK293 Cells , Signal Transduction/physiology , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Peptides , Glucagon-Like Peptide-1 Receptor/metabolism
2.
Article in English | MEDLINE | ID: mdl-38984948

ABSTRACT

The secretin-like, class B1 sub-family of seven transmembrane-spanning G protein coupled receptors (GPCRs) consists of 15 members that coordinate important physiological processes. These receptors bind peptide ligands and utilize a distinct mechanism of activation that is driven by evolutionarily conserved structural features. For the class B1 receptors, the C-terminus of the cognate ligand is initially recognized by the receptor via a large N-terminal extracellular domain that forms a hydrophobic ligand binding groove. This binding enables the N-terminus of the ligand to engage deep into a large volume, open transmembrane pocket of the receptor. Importantly, the phylogenetic basis of this ligand-receptor activation mechanism has provided opportunities to engineer analogues of several class B1 ligands for therapeutic use. Among the most successful of these are drugs targeting the glucagon-like peptide-1 (GLP-1) receptor for the treatment of type 2 diabetes and obesity. Recently, multi-functional agonists possessing activity at the GLP-1 receptor and the glucose-dependent insulinotropic polypeptide (GIP) receptor, such as tirzepatide, and others that also contain glucagon receptor activity, have been developed. In this article, we review members of the class B1 GPCR family with focus on receptors for GLP-1, GIP, and glucagon, including their signal transduction and receptor trafficking characteristics. The metabolic importance of these receptors is also highlighted, along with the benefit of poly-pharmacologic ligands. Further, key structural features and comparative analyses of high-resolution cryogenic electron microscopy structures for these receptors in active-state complex with either native ligands or multi-functional agonists are provided, supporting the pharmacological basis of such therapeutic agents.

3.
Ann Pharmacother ; 58(4): 398-406, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37522468

ABSTRACT

OBJECTIVE: This is a narrative review of incretin analogs and their effect on weight management in adult without diabetes. DATA SOURCES: Randomized controlled trials were identified by English language. PubMed/MEDLINE, Scopus, and Embase databases were searched from inception through June 2023 to identify all pertinent trials reporting outcomes on efficacy and safety search using the terms: tirzepatide, semaglutide, liraglutide, and obesity. STUDY SELECTION AND DATA EXTRACTION: Selected studies were included if the study population was composed of adults without diabetes being treated by glucagon-like peptide 1 (GLP-1) receptor agonists or glucose-dependent insulinotropic polypeptide (GIP)/GLP-1 agonists for weight management, and weight loss was assessed as a primary outcome. DATA SYNTHESIS: Fifteen studies involving 3 pharmacotherapies (liraglutide, semaglutide, and tirzepatide) were identified. Efficacy data supporting the use of these agents for weight management were promising when compared to placebo and/or other behavioral therapies. Percent weight loss ranged from 5.7% to 11.8%, 14.9% to 17.4%, and 15% to 20.9% for liraglutide, semaglutide, and tirzepatide, respectively. Safety data were relatively similar across all trials and identified gastrointestinal adverse effects as most common. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: Glucagon-like peptide 1 agonists are preferred for overweight or obese patients by the American Gastroenterological Association. Future guidelines may address tirzepatides' place in therapy as new evidence comes forth. Providers should consider patient-specific factors such as cost, adverse effects, drug interactions, and comorbidities when prescribing these agents and provide education regarding the need for concurrent diet and exercise modifications. CONCLUSIONS: All incretin analogs in this review are superior to placebo when used for weight management in adults without diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Incretins , Adult , Humans , Incretins/adverse effects , Liraglutide/adverse effects , Hypoglycemic Agents/adverse effects , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/therapeutic use , Obesity/drug therapy , Obesity/epidemiology , Weight Loss , Glucagon-Like Peptide-1 Receptor/agonists
4.
Int J Mol Sci ; 24(11)2023 May 25.
Article in English | MEDLINE | ID: mdl-37298217

ABSTRACT

The glucose-dependent insulinotropic polypeptide receptor (GIPR) is aberrantly expressed in about one-third of GH-secreting pituitary adenomas (GH-PAs) and has been associated with a paradoxical increase of GH after a glucose load. The reason for such an overexpression has not yet been clarified. In this work, we aimed to evaluate whether locus-specific changes in DNA methylation patterns could contribute to this phenomenon. By cloning bisulfite-sequencing PCR, we compared the methylation pattern of the GIPR locus in GIPR-positive (GIPR+) and GIPR-negative (GIPR-) GH-PAs. Then, to assess the correlation between Gipr expression and locus methylation, we induced global DNA methylation changes by treating the lactosomatotroph GH3 cells with 5-aza-2'-deoxycytidine. Differences in methylation levels were observed between GIPR+ and GIPR- GH-PAs, both within the promoter (31.9% vs. 68.2%, p < 0.05) and at two gene body regions (GB_1 20.7% vs. 9.1%; GB_2 51.2% vs. 65.8%, p < 0.05). GH3 cells treated with 5-aza-2'-deoxycytidine showed a ~75% reduction in Gipr steady-state level, possibly associated with the observed decrease in CpGs methylation. These results indicate that epigenetic regulation affects GIPR expression in GH-PAs, even though this possibly represents only a part of a much more complex regulatory mechanism.


Subject(s)
Adenoma , Growth Hormone-Secreting Pituitary Adenoma , Receptors, Gastrointestinal Hormone , Humans , Adenoma/genetics , Adenoma/metabolism , Decitabine , DNA Methylation , Epigenesis, Genetic , Growth Hormone-Secreting Pituitary Adenoma/genetics , Receptors, Gastrointestinal Hormone/metabolism
5.
J Biol Chem ; 295(33): 11529-11541, 2020 08 14.
Article in English | MEDLINE | ID: mdl-32554468

ABSTRACT

The insulinotropic actions of glucagon-like peptide 1 receptor (GLP-1R) in ß-cells have made it a useful target to manage type 2 diabetes. Metabolic stress reduces ß-cell sensitivity to GLP-1, yet the underlying mechanisms are unknown. We hypothesized that Glp1r expression is heterogeneous among ß-cells and that metabolic stress decreases the number of GLP-1R-positive ß-cells. Here, analyses of publicly available single-cell RNA-Seq sequencing (scRNASeq) data from mouse and human ß-cells indicated that significant populations of ß-cells do not express the Glp1r gene, supporting heterogeneous GLP-1R expression. To check these results, we used complementary approaches employing FACS coupled with quantitative RT-PCR, a validated GLP-1R antibody, and flow cytometry to quantify GLP-1R promoter activity, gene expression, and protein expression in mouse α-, ß-, and δ-cells. Experiments with Glp1r reporter mice and a validated GLP-1R antibody indicated that >90% of the ß-cells are GLP-1R positive, contradicting the findings with the scRNASeq data. α-cells did not express Glp1r mRNA and δ-cells expressed Glp1r mRNA but not protein. We also examined the expression patterns of GLP-1R in mouse models of metabolic stress. Multiparous female mice had significantly decreased ß-cell Glp1r expression, but no reduction in GLP-1R protein levels or GLP-1R-mediated insulin secretion. These findings suggest caution in interpreting the results of scRNASeq for low-abundance transcripts such as the incretin receptors and indicate that GLP-1R is widely expressed in ß-cells, absent in α-cells, and expressed at the mRNA, but not protein, level in δ-cells.


Subject(s)
Glucagon-Like Peptide-1 Receptor/genetics , Insulin-Secreting Cells/metabolism , Animals , Cells, Cultured , Gene Expression , Glucagon-Like Peptide-1 Receptor/analysis , Humans , Mice , Mice, Inbred C57BL , Single-Cell Analysis
6.
Biochem Biophys Res Commun ; 534: 121-127, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33321289

ABSTRACT

The intrinsic capacity of axonal growth is varied among the neurons form different tissues or different developmental stages. In this study, we established an in vitro model to compare the axonal growth of neurons from embryonic 18 days, post-natal 1 day and post-natal 3 days rat. The E18 neurons showed powerful ability of neuritogenensis and axon outgrowth and the ability decreased rapidly along with development. The transcriptome profile of these neurons revealed a set of genes positively correlated with the capacity of neurite outgrowth. Glucose-dependent insulinotropic polypeptide receptor (GIPR) is identified as a gene to promote neurite outgrowth, which was approved by siRNA knock down assay in E18 neuron. Glucose-dependent insulinotropic polypeptide (GIP), a ligand of GIPR secreted from enteroendocrine K cells, is well-known for its role in nutrient sensing and intake. To verify the effect of GIP-GIPR signal on neurite outgrowth, we administrated GIP to stimulate the E18 neurons, the results showed that GIP significantly improved extension of axon. We further revealed that GIP increased Rac1/Cdc42 phosphorylation in Akt dependent manner. In summary, our study established an in vitro model to screen the genes involved in neurite outgrowth, and we provided mechanical insight on the GIP-GIPR axis to promote axonal outgrowth.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Neuronal Outgrowth/physiology , Neurons/physiology , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Animals , Animals, Newborn , Axons/physiology , Cells, Cultured , Cerebral Cortex/cytology , Cerebral Cortex/embryology , Cerebral Cortex/growth & development , Female , Gastric Inhibitory Polypeptide/genetics , Gene Expression Regulation, Developmental , Neurons/cytology , Rats, Sprague-Dawley , Receptors, Gastrointestinal Hormone/genetics
7.
Rev Endocr Metab Disord ; 21(1): 165-183, 2020 03.
Article in English | MEDLINE | ID: mdl-31933128

ABSTRACT

The glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone produced in the gastrointestinal tract in response to nutrients. GIP has a variety of effects on different systems, including the potentiation of insulin secretion from pancreatic ß-cells after food intake (i.e. incretin effect), which is probably the most important. GIP effects are mediated by the GIP receptor (GIPR), a G protein-coupled receptor expressed in several tissues, including islet ß-cells, adipocytes, bone cells, and brain. As well as its involvement in metabolic disorders (e.g. it contributes to the impaired postprandial insulin secretion in type 2 diabetes (T2DM), and to the pathogenesis of obesity and associated insulin resistance), an inappropriate GIP/GIPR axis activation of potential diagnostic and prognostic value has been reported in several endocrine tumors in recent years. The ectopic GIPR expression seen in patients with overt Cushing syndrome and primary bilateral macronodular adrenal hyperplasia or unilateral cortisol-producing adenoma has been associated with an inverse rhythm of cortisol secretion, with low fasting morning plasma levels that increase after eating. On the other hand, most acromegalic patients with an unusual GH response to oral glucose suppression have GIPR-positive somatotropinomas, and a milder phenotype, and are more responsive to medical treatment. Neuroendocrine tumors are characterized by a strong GIPR expression that may correlate positively or inversely with the proliferative index MIB-1, and that seems an attractive target for developing novel radioligands. The main purpose of this review is to summarize the role of the GIP/GIPR axis in endocrine neoplasia, in the experimental and the clinical settings.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Neuroendocrine Tumors/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Adenoma/metabolism , Diabetes Mellitus, Type 2 , Gene Expression Regulation, Neoplastic , Growth Hormone-Secreting Pituitary Adenoma/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Receptors, Gastrointestinal Hormone/genetics
8.
Diabetes Obes Metab ; 21(5): 1168-1176, 2019 05.
Article in English | MEDLINE | ID: mdl-30784161

ABSTRACT

AIMS: Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone that augments insulin secretion in pancreatic ß-cells via its glucose-dependent insulinotropic polypeptide receptor (GIPR). Recent genome-wide association studies identified a single nucleotide variant (SNV) in the GIPR encoding gene (GIPR), rs1800437, that is associated with obesity and insulin resistance. In the present study, we tested whether GIPR variants contribute to obesity and disturb glucose homeostasis or diabetes in specific patient populations. MATERIALS AND METHODS: Exon sequencing of GIPR was performed in 164 children with obesity and insulin resistance and in 80 children with paediatric-onset diabetes of unknown origin. The Study of Health in Pomerania (SHIP) cohort, comprising 8320 adults, was screened for the GIPR variant Arg217Leu. GIPR variants were expressed in COS-7 cells and cAMP production was measured upon stimulation with GIP. Cell surface expression was determined by ELISA. Protein homology modelling of the GIPR variants was performed to extract three-dimensional information of the receptor. RESULTS: A heterozygous missense GIPR variant Arg217Leu (rs200485112) was identified in a patient of Asian ancestry. Functional characterization of Arg217Leu revealed reduced surface expression and signalling after GIP challenge. The homology model of the GIPR structure supports the observed functional relevance of Arg217Leu. CONCLUSION: In vitro functional studies and protein homology modelling indicate a potential relevance of the GIPR variant Arg217Leu in receptor function. The heterozygous variant displayed partial co-segregation with diabetes. Based on these findings, we suggest that GIPR variants may play a role in disturbed glucose homeostasis and may be of clinical relevance in homozygous patients.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Polymorphism, Single Nucleotide , Receptors, Gastrointestinal Hormone/genetics , Adolescent , Age of Onset , Amino Acid Substitution/genetics , Animals , Arginine/genetics , COS Cells , Child , Chlorocebus aethiops , Cohort Studies , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/metabolism , Female , Gene Frequency , Genome-Wide Association Study , Germany/epidemiology , Homozygote , Humans , Insulin Resistance/genetics , Leucine/genetics , Male , Pediatric Obesity/complications , Pediatric Obesity/epidemiology , Pediatric Obesity/genetics
9.
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
10.
Nucl Med Biol ; 128-129: 108876, 2024.
Article in English | MEDLINE | ID: mdl-38241936

ABSTRACT

BACKGROUND: The beneficial role of glucose-dependent insulinotropic polypeptide receptor (GIPR) in weight control and maintaining glucose levels has led to the development of several multi-agonistic peptide drug candidates, targeting GIPR and glucagon like peptide 1 receptor (GLP1R) and/or the glucagon receptor (GCGR). The in vivo quantification of target occupancy by these drugs would accelerate the development of new drug candidates. The aim of this study was to evaluate a novel peptide (GIP1234), based on previously reported ligand DOTA-GIP-C803, modified with a fatty acid moiety to prolong its blood circulation. It would allow higher target tissue exposure and consequently improved peptide uptake as well as in vivo PET imaging and quantification of GIPR occupancy by novel drugs of interest. METHOD: A 40 amino acid residue peptide (GIP1234) was synthesized based on DOTA-GIP-C803, in turn based on the sequences of endogenous GIP and Exendin-4 with specific amino acid modifications to obtain GIPR selectivity. A palmitoyl fatty acid chain was furthermore added at Lys14 via a glutamic acid linker to prolong its blood circulation time by the interaction with albumin. GIP1234 was conjugated with a DOTA chelator at the C-terminal cysteine residue to achieve 68Ga radiolabeling. The resulting PET probe, [68Ga]Ga-DOTA-GIP1234 was evaluated for receptor binding specificity and selectivity using HEK293 cells transfected with human GIPR, GLP1R, or GCGR. Blocking experiments with tirzepatide (2 µM) were conducted using huGIPR HEK293 cells to investigate binding specificity. Ex vivo and in vivo organ distribution of [68Ga]Ga-DOTA-GIP1234 was studied in rats and a pig in comparison to [68Ga]Ga-DOTA-C803-GIP. Binding of [68Ga]Ga-DOTA-GIP1234 to albumin was assessed in situ using polyacrylamide gel electrophoresis (PAGE). The stability was tested in formulation buffer and rat blood plasma. RESULTS: [68Ga]Ga-DOTA-GIP1234 was synthesized with non-decay corrected radiochemical yield of 88 ± 3.7 % and radiochemical purity of 97.8 ± 0.8 %. The molar activity for the radiotracer was 8.1 ± 1.1 MBq/nmol. [68Ga]Ga-DOTA-GIP1234 was stable and maintained affinity to huGIPR HEK293 cells (dissociation constant (Kd) = 40 ± 12.5 nM). The binding of [68Ga]Ga-DOTA-GIP1234 to huGCGR and huGLP1R cells was insignificant. Pre-incubation of huGIPR HEK293 cell sections with tirzepatide resulted in the decrease of [68Ga]Ga-DOTA-GIP1234 binding by close to 90 %. [68Ga]Ga-DOTA-GIP1234 displayed slow blood clearance in pigs with SUV = 3.5 after 60 min. Blood retention of the tracer in rat was 2-fold higher than that of [68Ga]Ga-DOTA-C803-GIP. [68Ga]Ga-DOTA-GIP1234 also demonstrated strong liver uptake in both pig and rat combined with decreased renal excretion. The concentration dependent binding of [68Ga]Ga-DOTA-GIP1234 to albumin was confirmed in situ by PAGE. CONCLUSION: [68Ga]Ga-DOTA-GIP1234 demonstrated nanomolar affinity and selectivity for huGIPR in vitro. Addition of a fatty acid moiety prolonged blood circulation time and tissue exposure in both rat and pig in vivo. However, the liver uptake was also increased which may make PET imaging of abdominal tissues such as pancreas challenging. The investigation of the influence of fatty acid moiety on the biological performance of the peptide ligand paved the way for further rational design of GIPR ligand analogues with improved characteristics.


Subject(s)
Gallium Radioisotopes , Peptides , Receptors, Gastrointestinal Hormone , Rats , Humans , Animals , Swine , HEK293 Cells , Ligands , Gallium Radioisotopes/chemistry , Half-Life , Peptides/chemistry , Albumins , Amino Acids
11.
Dig Liver Dis ; 56(10): 1730-1737, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38735797

ABSTRACT

OBJECTIVE: Non-alcoholic fatty liver disease (NAFLD) is becoming the most common chronic liver disease worldwide while still lacks drugs for treatment or prevention. We aimed to investigate the causal role of glucose-dependent insulinotropic polypeptide receptor agonists (GIPRAs) on NAFLD and identify the mediated risk factors by which GIPRAs exert their therapeutic effects. METHODS: Genetic proxies of GIPRAs were identified as cis-SNPs of GIPR associated with both the gene expression level and HbA1c and analyses including colocalization and linkage disequilibrium (LD) were performed for validation. We then performed two-sample two-step mendelian randomization to determine the causal effect of GIPRAs on NAFLD. RESULTS: The MR analysis suggested genetic proxies of GIPRAs were causally associated with reduced risk of NAFLD (Odds ratio (OR): 0.46, 95 % confidence interval (95 % CI): 0.24-0.88, P = 0.02) and T2DM (OR: 0.10, 95 % CI: 0.07-0.13, P < 0.01). In addition, Mediation analysis showed evidence of indirect effect of GIPRAs on NAFLD via TRIG (0.88, [0.85-0.92], P < 0.01) and HDL-C (0.85, [0.80-0.90], P < 0.01). CONCLUSIONS: Our study provided strong evidence to support the causal role of GIPRAs on reducing the risk of NAFLD probably through improving lipid metabolism, especially TG and HDL-C, providing guidance for future clinical trials.


Subject(s)
Non-alcoholic Fatty Liver Disease , Receptors, Gastrointestinal Hormone , Humans , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/drug therapy , Glycated Hemoglobin , Linkage Disequilibrium , Mediation Analysis , Mendelian Randomization Analysis , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/drug therapy , Polymorphism, Single Nucleotide , Receptors, Gastrointestinal Hormone/genetics
12.
J Endocrinol ; 263(1)2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39013412

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like-peptide-1 (GLP-1) are incretin hormones that stimulate insulin secretion and improve glycemic control in individuals with type 2 diabetes (T2D). Data from several cardiovascular outcome trials for GLP-1 receptor (GLP-1R) agonists have demonstrated significant reductions in the occurrence of major adverse cardiovascular events in individuals with T2D. Although the cardiovascular actions attributed to GLP-1R agonism have been extensively studied, little is known regarding the cardiovascular consequences attributed to GIP receptor (GIPR) agonism. As there is now an increasing focus on the development of incretin-based co-agonist therapies that activate both the GLP-1R and GIPR, it is imperative that we understand the mechanism(s) through which these incretins impact cardiovascular function. This is especially important considering that cardiovascular disease represents the leading cause of death in individuals with T2D. With increasing evidence that perturbations in cardiac energy metabolism are a major contributor to the pathology of diabetes-related cardiovascular disease, this may represent a key component through which GLP-1R and GIPR agonism influence cardiovascular outcomes. Not only do GIP and GLP-1 increase the secretion of insulin, they may also modify glucagon secretion, both of which have potent actions on cardiac substrate utilization. Herein we will discuss the potential direct and indirect actions through which GLP-1R and GIPR agonism impact cardiac energy metabolism while interrogating the evidence to support whether such actions may account for incretin-mediated cardioprotection in T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Energy Metabolism , Incretins , Humans , Incretins/therapeutic use , Incretins/pharmacology , Incretins/metabolism , Energy Metabolism/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Heart/drug effects , Cardiovascular Diseases/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/agonists , Glucagon-Like Peptide 1/metabolism , Myocardium/metabolism
13.
J Diabetes Investig ; 14(2): 189-192, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36373430

ABSTRACT

Mechanisms of dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists and glucagon-like peptide-1 receptor/glucose-dependent insulinotropic polypeptide receptor dual-agonist in glycemic control and/or weight loss.


Subject(s)
Diabetes Mellitus, Type 2 , Dipeptidyl-Peptidase IV Inhibitors , Humans , Incretins/therapeutic use , Incretins/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/pharmacology
14.
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
15.
Ther Clin Risk Manag ; 18: 955-964, 2022.
Article in English | MEDLINE | ID: mdl-36199834

ABSTRACT

Tirzepatide is a novel "twincretin" with glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide receptor agonist activity, which was recently approved by the Food and Drug Administration for the treatment of type 2 diabetes mellitus. In this review, we discuss preclinical and mechanistic human studies, which demonstrate improvements in insulin sensitivity and beta-cell function with the use of tirzepatide, as compared to placebo and glucagon-like peptide 1 receptor agonists. We then discuss SURPASS trials 1-5, which evaluated the safety and efficacy of tirzepatide for type 2 diabetes mellitus as either monotherapy or combination therapy with other antidiabetic agents. The magnitude of tirzepatide's effects and the efficacy relative to other anti-diabetes medications on weight, glycemic control, and beta-cell function may prove beneficial for the treatment of early type 2 diabetes mellitus. Further studies, including data on cardiovascular outcomes and long-term safety, will continue to elucidate the role of tirzepatide in the treatment algorithm of type 2 diabetes mellitus.

16.
J Cardiovasc Pharmacol Ther ; 27: 10742484221146371, 2022.
Article in English | MEDLINE | ID: mdl-36546652

ABSTRACT

The obesity pandemic is accompanied by increased risk of developing metabolic syndrome (MetS) and related conditions: non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH), type 2 diabetes mellitus (T2DM) and cardiovascular (CV) disease (CVD). Lifestyle, as well as an imbalance of energy intake/expenditure, genetic predisposition, and epigenetics could lead to a dysmetabolic milieu, which is the cornerstone for the development of cardiometabolic complications. Glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) and dual glucose-dependent insulinotropic polypeptide (GIP)/GLP-1 RAs promote positive effects on most components of the "cardiometabolic continuum" and consequently help reduce the need for polypharmacy. In this review, we highlight the main pathophysiological mechanisms and risk factors (RFs), that could be controlled by GLP-1 and dual GIP/GLP-1 RAs independently or through synergism or differences in their mode of action. We also address the evidence on the use of GLP-1 and dual GIP/GLP-1 RAs in the treatment of obesity, MetS and its related conditions (prediabetes, T2DM and NAFLD/NASH). In conclusion, GLP-1 RAs have already been established for the treatment of T2DM, obesity and cardioprotection in T2DM patients, while dual GIP/GLP-1 RAs appear to have the potential to possibly surpass them for the same indications. However, their use in the prevention of T2DM and the treatment of complex cardiometabolic metabolic diseases, such as NAFLD/NASH or other metabolic disorders, would benefit from more evidence and a thorough clinical patient-centered approach. There is a need to identify those patients in whom the metabolic component predominates, and whether the benefits outweigh any potential harm.


Subject(s)
Diabetes Mellitus, Type 2 , Metabolic Syndrome , Non-alcoholic Fatty Liver Disease , Prediabetic State , Humans , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/epidemiology , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/complications , Glucagon-Like Peptide-1 Receptor/agonists , Metabolic Syndrome/drug therapy , Metabolic Syndrome/complications , Prediabetic State/diagnosis , Prediabetic State/drug therapy , Prediabetic State/complications , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glucagon-Like Peptide 1/therapeutic use , Obesity/drug therapy , Obesity/complications , Glucose/therapeutic use , Peptides/therapeutic use
17.
Biomed Pharmacother ; 142: 112063, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34463265

ABSTRACT

In the present study, we investigated the effects of Galla Rhois (GR) on obesity and gene expression. We prepared a GR extract and various solvent fractions and evaluated the degree to which they inhibited adipocyte differentiation and adipogenesis in vitro. Among them, the GR ethyl acetate fraction (GE) had the lowest EC50 for adipocyte differentiation and adipogenesis and thus was selected for in vivo experiments. We induced obesity in C57BL/6 mice by providing them a high-fat diet (HFD). Then, GE (10-40 mg/kg) or orlistat (positive control, 4 mg/kg) was orally administered daily for six weeks. Mean body weights and weight gain were significantly lower in the GE40 group (40 mg/kg of GE) compared with the HFD group (p < 0.05). The most significant changes in serum glucose, total cholesterol, and triglyceride levels were confirmed in the GE40 group (p < 0.05). Epididymal fat was weighed and stained for body fat measurement, and significant differences were recorded from GE10 to GE40 (p < 0.05). Finally, 3T3-L1 pre-adipocytes were treated with GE, and cDNA from these cells was used for microarray analysis and qRT-PCR. Microarray analysis revealed 13 genes up-regulated and 21 genes down-regulated by GE. From the qRT-PCR analysis, we found that GE altered the mRNA expression of eosinophil peroxidase, glucose-dependent insulinotropic polypeptide receptor, and apolipoprotein B. Based on this study, we suggest that GR could be developed as an anti-obesity therapeutic agent.


Subject(s)
Adipogenesis/drug effects , Anti-Obesity Agents/pharmacology , Biological Products/pharmacology , Obesity/drug therapy , 3T3-L1 Cells , Adipocytes/cytology , Adipocytes/drug effects , Adipogenesis/genetics , Animals , Body Weight/drug effects , Cell Differentiation/drug effects , Diet, High-Fat , Gene Expression Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Weight Gain/drug effects
18.
Biochem Pharmacol ; 192: 114715, 2021 10.
Article in English | MEDLINE | ID: mdl-34339714

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and its receptor (GIPR) are part of the incretin system that regulates glucose homeostasis. A series of GIPR residues putatively important for ligand binding and receptor activation were mutated and pharmacologically evaluated using GIPR selective agonists in cAMP accumulation, ERK1/2 phosphorylation (pERK1/2) and ß-arrestin 2 recruitment assays. The impact of mutation on ligand efficacy was determined by operational modelling of experimental data for each mutant, with results mapped onto the full-length, active-state GIPR structure. Two interaction networks, comprising transmembrane helix (TM) 7, TM1 and TM2, and extracellular loop (ECL) 2, TM5 and ECL3 were revealed, respectively. Both networks were critical for Gαs-mediated cAMP accumulation and the recruitment of ß-arrestin 2, however, cAMP response was more sensitive to alanine substitution, with most mutated residues displaying reduced signaling. Unlike the other two assays, activation of ERK1/2 was largely independent of the network involving ECL2, TM5 and ECL3, indicating that pERK1/2 is at least partially distinct from Gαs or ß-arrestin pathways and this network is also crucial for potential biased agonism at GIPR. Collectively, our work advances understanding of the structure-function relationship of GIPR and provides a framework for the design and/or interpretation of GIP analogues with unique signaling profiles.


Subject(s)
Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Amino Acid Sequence , Animals , Binding Sites/drug effects , Binding Sites/physiology , COS Cells , Chlorocebus aethiops , Dose-Response Relationship, Drug , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/pharmacology , HEK293 Cells , Humans , Mutation/physiology , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , Protein Structure, Secondary , Receptors, Gastrointestinal Hormone/chemistry
19.
Elife ; 102021 07 13.
Article in English | MEDLINE | ID: mdl-34254582

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) is a peptide hormone that exerts crucial metabolic functions by binding and activating its cognate receptor, GIPR. As an important therapeutic target, GIPR has been subjected to intensive structural studies without success. Here, we report the cryo-EM structure of the human GIPR in complex with GIP and a Gs heterotrimer at a global resolution of 2.9 Å. GIP adopts a single straight helix with its N terminus dipped into the receptor transmembrane domain (TMD), while the C terminus is closely associated with the extracellular domain and extracellular loop 1. GIPR employs conserved residues in the lower half of the TMD pocket to recognize the common segments shared by GIP homologous peptides, while uses non-conserved residues in the upper half of the TMD pocket to interact with residues specific for GIP. These results provide a structural framework of hormone recognition and GIPR activation.


Subject(s)
Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Animals , Cell Line , Cryoelectron Microscopy , Humans , Ligands , Molecular Dynamics Simulation , Protein Conformation , Protein Conformation, alpha-Helical , Protein Domains
20.
Front Immunol ; 12: 643144, 2021.
Article in English | MEDLINE | ID: mdl-33717200

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) communicates information on energy availability from the gut to peripheral tissues. Disruption of its signaling in myeloid immune cells during high-fat diet (HFD)-induced obesity impairs energy homeostasis due to the unrestrained metabolically deleterious actions of S100A8/A9 alarmin. White adipose tissue (WAT) type 2 immune cell networks are important for maintaining metabolic and energy homeostasis and limiting obesity-induced inflammation. Nevertheless, the consequences of losing immune cell GIP receptor (GIPR) signaling on type 2 immunity in WAT remains unknown. Bone marrow (BM) chimerism was used to generate mice with GIPR (Gipr-/- BM) and GIPR/S100A8/A9 (Gipr-/- /S100a9-/- BM) deletion in immune cells. These mice were subjected to short (5 weeks) and progressive (14 weeks) HFD regimens. GIPR-deficiency was also targeted to myeloid cells by crossing Giprfl/fl mice and Lyz2cre/+ mice (LysMΔGipr ). Under both short and progressive HFD regimens, Gipr-/- BM mice exhibited altered expression of key type 2 immune cytokines in the epididymal visceral WAT (epiWAT), but not in subcutaneous inguinal WAT. This was further linked to declined representation of type 2 immune cells in epiWAT, such as group 2 innate lymphoid cells (ILC2), eosinophils, and FOXP3+ regulatory T cells (Tregs). Co-deletion of S100A8/A9 in Gipr-/- immune cells reversed the impairment of type 2 cytokine expression in epiWAT, suggesting a mechanistic role for this alarmin in type 2 immune suppression. LysMΔGipr mice on HFD also displayed altered expression of type 2 immune mediators, highlighting that GIPR-deficiency in myeloid immune cells is responsible for the impairment of type 2 immune networks. Finally, abrogated GIPR signaling in immune cells also affected adipocyte fraction cells, inducing their increased production of the beiging interfering cytokine IL-10 and stress- related type 2 cytokine IL-13. Collectively, these findings attribute an important role for GIPR in myeloid immune cells in supporting WAT type 2 immunity.


Subject(s)
Adipose Tissue, White/immunology , Lymphocytes/immunology , Obesity/immunology , Receptors, Gastrointestinal Hormone/physiology , Adipose Tissue, White/metabolism , Animals , Calgranulin A/physiology , Calgranulin B/physiology , Diet, High-Fat , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Signal Transduction/physiology , Thermogenesis
SELECTION OF CITATIONS
SEARCH DETAIL