Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.057
Filter
1.
Mol Biol Rep ; 51(1): 966, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39240462

ABSTRACT

In humans, 15 genes encode the class B1 family of GPCRs, which are polypeptide hormone receptors characterized by having a large N-terminal extracellular domain (ECD) and receive signals from outside the cell to activate cellular response. For example, the insulinotropic polypeptide (GIP) stimulates the glucose-dependent insulinotropic polypeptide receptor (GIPR), while the glucagon receptor (GCGR) responds to glucagon by increasing blood glucose levels and promoting the breakdown of liver glycogen to induce the production of insulin. The glucagon-like peptides 1 and 2 (GLP-1 and GLP-2) elicit a response from glucagon-like peptide receptor types 1 and 2 (GLP1R and GLP2R), respectively. Since these receptors are implicated in the pathogenesis of diabetes, studying their activation is crucial for the development of effective therapies for the condition. With more structural information being revealed by experimental methods such as X-ray crystallography, cryo-EM, and NMR, the activation mechanism of class B1 GPCRs becomes unraveled. The available crystal and cryo-EM structures reveal that class B1 GPCRs follow a two-step model for peptide binding and receptor activation. The regions close to the C-termini of hormones interact with the N-terminal ECD of the receptor while the regions close to the N-terminus of the peptide interact with the TM domain and transmit signals. This review highlights the structural details of class B1 GPCRs and their conformational changes following activation. The roles of MD simulation in characterizing those conformational changes are briefly discussed, providing insights into the potential structural exploration for future ligand designs.


Subject(s)
Receptors, G-Protein-Coupled , Humans , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Crystallography, X-Ray/methods , Protein Conformation , Animals , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/genetics , Glucagon-Like Peptide 1/metabolism , Models, Molecular , Protein Binding , Signal Transduction , Receptors, Glucagon/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/chemistry
2.
Mol Metab ; 89: 102024, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39236784

ABSTRACT

OBJECTIVE: Glucagon has long been proposed as a component of multi-agonist obesity therapeutics due to its ability to induce energy expenditure and cause weight loss. However, chronic glucagon-receptor agonism has been associated with a reduction in circulating amino acids and loss of lean mass. Importantly, it is currently not known whether the metabolic benefits of glucagon can be maintained under contexts that allow the defence of lean mass. METHODS: We investigate the metabolic effects of the long-acting glucagon receptor agonist, G108, when administered to obese mice at low-doses, and with dietary protein supplementation. RESULTS: Dietary protein supplementation can only fully defend lean mass at a low dose of G108 that is sub-anorectic and does not reduce fat mass. However, in this context, G108 is still highly effective at improving glucose tolerance and reducing liver fat in obese mice. Mechanistically, liver RNA-Seq analysis reveals that dietary protein supplementation defends anabolic processes in low-dose G108-treated mice, and its effects on treatment-relevant glucose and lipid pathways are preserved. CONCLUSION: Glucagon-mediated energy expenditure and weight loss may be mechanistically coupled to hypoaminocidemia and lean mass loss. However, our data suggest that glucagon can treat MAFLD at doses which allow full defence of lean mass given sufficient dietary protein intake. Therefore, proportionate glucagon therapy may be safe and effective in targeting hepatocytes and improving in glycaemia and liver fat.


Subject(s)
Dietary Proteins , Energy Metabolism , Glucagon , Mice, Inbred C57BL , Obesity , Receptors, Glucagon , Animals , Mice , Receptors, Glucagon/metabolism , Receptors, Glucagon/agonists , Male , Glucagon/metabolism , Obesity/metabolism , Obesity/drug therapy , Energy Metabolism/drug effects , Dietary Proteins/pharmacology , Dietary Proteins/metabolism , Liver/metabolism , Mice, Obese , Weight Loss/drug effects
3.
PLoS One ; 19(8): e0302565, 2024.
Article in English | MEDLINE | ID: mdl-39196987

ABSTRACT

Obesity is a growing concern. 42.3% of people in the U.S were considered obese between 2017-2018. Much is still unknown about the genetic components that contribute to weight gain. In humans, the hormone glucagon is a major contributor to the body's energy regulation as it signals for the breakdown of lipids. Treatments targeting the glucagon pathway have helped patients with both weight loss and appetite suppression. Understanding the genetic modifiers of glucagon signaling and its downstream pathways could enable the development of a wider variety of effective therapeutics. In this study, we blocked the glucagon pathway in Drosophila melanogaster by reducing the expression of the fly ortholog of the glucagon receptor (AKHR). We then crossed our model to the Drosophila Genetic Reference Panel (DGRP) and looked for natural variation in fat content. We used variation in larval density to identify candidate modifier genes through a genome-wide association study. We then tested these modifier genes by increasing or decreasing their expression in the AKHR model. We screened these candidates initially with the same density assay used in the original study to narrow down to four candidate genes that substantially impacted the density of the larvae: THADA, AmyD, GluRIIC, and CG9826. We further characterized these candidates using biochemical assays to analyze stored metabolites such as triglycerides, glucose, glycogen, and protein under control, high sugar, and high fat conditions to see if the larvae are resistant to environmental changes. Our results indicate consistency between the results of the density assay and direct measurement of metabolite levels. In particular, THADA and AmyD are highlighted as interesting genes for additional study. We hope to improve our understanding of the glucagon signaling pathway, obesity, and lipid metabolism. We also aim to provide candidate genes that can be regarded as future therapeutic targets.


Subject(s)
Drosophila Proteins , Drosophila melanogaster , Glucagon , Larva , Receptors, Glucagon , Signal Transduction , Animals , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Glucagon/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Larva/genetics , Larva/metabolism , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Genome-Wide Association Study , Male , Obesity/genetics , Obesity/metabolism , Female
4.
J Med Chem ; 67(17): 14820-14839, 2024 Sep 12.
Article in English | MEDLINE | ID: mdl-39140772

ABSTRACT

Various small molecule GLP1R agonists have been developed and tested for treating type 2 diabetes (T2DM) and obesity. However, many of these new compounds have drawbacks, such as potential hERG inhibition, lower activity compared to natural GLP-1, limited oral bioavailability in cynomolgus monkeys, and short duration of action. Recently, a new category of 3-phenyloxetane derivative GLP1R agonists with enhanced hERG inhibition has been discovered. Using an AIDD/CADD method, compound 14 (DD202-114) was identified as a potent and selective GLP1R agonist, which was chosen as a preclinical candidate (PCC). Compound 14 demonstrates full agonistic efficacy in promoting cAMP accumulation and possesses favorable drug-like characteristics compared to the clinical drug candidate Danuglipron. Additionally, in hGLP-1R knock-in mice, compound 14 displayed a sustained pharmacological effect, effectively reducing blood glucose levels and food intake. These findings suggest that compound 14 holds promise as a future treatment option for T2DM and obesity, offering improved properties.


Subject(s)
Drug Design , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Animals , Humans , Mice , Structure-Activity Relationship , Male , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemical synthesis , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism
6.
Int J Mol Sci ; 25(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39125959

ABSTRACT

Glucagon receptor (GCGR) is a class B1 G-protein-coupled receptor that plays a crucial role in maintaining human blood glucose homeostasis and is a significant target for the treatment of type 2 diabetes mellitus (T2DM). Currently, six small molecules (Bay 27-9955, MK-0893, MK-3577, LY2409021, PF-06291874, and LGD-6972) have been tested or are undergoing clinical trials, but only the binding site of MK-0893 has been resolved. To predict binding sites for other small molecules, we utilized both the crystal structure of the GCGR and MK-0893 complex and dynamic conformations. We docked five small molecules and selected the best conformation based on binding mode, docking score, and binding free energy. We performed MD simulations to verify the binding mode of the selected small molecules. Moreover, when selecting conformations, results of competitive binding were referred to. MD simulation indicated that Bay 27-9955 exhibits moderate binding stability in Pocket 3. MK-3577, LY2409021, and PF-06291874 exhibited highly stable binding to Pocket 2, consistent with experimental results. However, LY2409021 may also bind to Pocket 5. Additionally, LGD-6972 exhibited relatively stable binding in Pocket 5. We also conducted structural modifications of LGD-6972 based on the results of MD simulations and predicted its analogues' bioavailability, providing a reference for the study of GCGR small molecules.


Subject(s)
Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Conformation , Receptors, Glucagon , Binding Sites , Humans , Crystallography, X-Ray , Receptors, Glucagon/chemistry , Receptors, Glucagon/metabolism , Protein Binding , Small Molecule Libraries/chemistry , Ligands , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism
7.
Biomolecules ; 14(7)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39062586

ABSTRACT

Glucagon-like peptide-1 (GLP-1)-based drugs have been approved by the United States Food and Drug Administration (FDA) and are widely used to treat type 2 diabetes mellitus (T2DM) and obesity. More recent developments of unimolecular peptides targeting multiple incretin-related receptors ("multi-agonists"), including the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) and the glucagon (Gcg) receptor (GcgR), have emerged with the aim of enhancing drug benefits. In this study, we utilized human and mouse microglial cell lines, HMC3 and IMG, respectively, together with the human neuroblastoma SH-SY5Y cell line as cellular models of neurodegeneration. Using these cell lines, we studied the neuroprotective and anti-inflammatory capacity of several multi-agonists in comparison with a single GLP-1 receptor (GLP-1R) agonist, exendin-4. Our data demonstrate that the two selected GLP-1R/GIPR dual agonists and a GLP-1R/GIPR/GcgR triple agonist not only have neurotrophic and neuroprotective effects but also have anti-neuroinflammatory properties, as indicated by the decreased microglial cyclooxygenase 2 (COX2) expression, nitrite production, and pro-inflammatory cytokine release. In addition, our results indicate that these multi-agonists have the potential to outperform commercially available single GLP-1R agonists in neurodegenerative disease treatment.


Subject(s)
Anti-Inflammatory Agents , Glucagon-Like Peptide-1 Receptor , Incretins , Neuroprotective Agents , Humans , Animals , Neuroprotective Agents/pharmacology , Neuroprotective Agents/chemistry , Incretins/pharmacology , Mice , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Cell Line , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism , Exenatide/pharmacology , Microglia/drug effects , Microglia/metabolism , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Cell Line, Tumor , Peptides/pharmacology , Peptides/chemistry , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/agonists
8.
Circ Res ; 135(5): 614-628, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39011638

ABSTRACT

BACKGROUND: Heart failure with preserved ejection fraction (HFpEF) is an emerging major unmet need and one of the most significant clinic challenges in cardiology. The pathogenesis of HFpEF is associated with multiple risk factors. Hypertension and metabolic disorders associated with obesity are the 2 most prominent comorbidities observed in patients with HFpEF. Although hypertension-induced mechanical overload has long been recognized as a potent contributor to heart failure with reduced ejection fraction, the synergistic interaction between mechanical overload and metabolic disorders in the pathogenesis of HFpEF remains poorly characterized. METHOD: We investigated the functional outcome and the underlying mechanisms from concurrent mechanic and metabolic stresses in the heart by applying transverse aortic constriction in lean C57Bl/6J or obese/diabetic B6.Cg-Lepob/J (ob/ob) mice, followed by single-nuclei RNA-seq and targeted manipulation of a top-ranked signaling pathway differentially affected in the 2 experimental cohorts. RESULTS: In contrast to the post-transverse aortic constriction C57Bl/6J lean mice, which developed pathological features of heart failure with reduced ejection fraction over time, the post-transverse aortic constriction ob/ob mice showed no significant changes in ejection fraction but developed characteristic pathological features of HFpEF, including diastolic dysfunction, worsened cardiac hypertrophy, and pathological remodeling, along with further deterioration of exercise intolerance. Single-nuclei RNA-seq analysis revealed significant transcriptome reprogramming in the cardiomyocytes stressed by both pressure overload and obesity/diabetes, markedly distinct from the cardiomyocytes singularly stressed by pressure overload or obesity/diabetes. Furthermore, glucagon signaling was identified as the top-ranked signaling pathway affected in the cardiomyocytes associated with HFpEF. Treatment with a glucagon receptor antagonist significantly ameliorated the progression of HFpEF-related pathological features in 2 independent preclinical models. Importantly, cardiomyocyte-specific genetic deletion of the glucagon receptor also significantly improved cardiac function in response to pressure overload and metabolic stress. CONCLUSIONS: These findings identify glucagon receptor signaling in cardiomyocytes as a critical determinant of HFpEF progression and provide proof-of-concept support for glucagon receptor antagonism as a potential therapy for the disease.


Subject(s)
Heart Failure , Mice, Inbred C57BL , Stroke Volume , Animals , Heart Failure/physiopathology , Heart Failure/metabolism , Heart Failure/drug therapy , Heart Failure/etiology , Stroke Volume/drug effects , Mice , Male , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/metabolism , Receptors, Glucagon/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Mice, Obese , Ventricular Function, Left/drug effects , Obesity/metabolism , Obesity/physiopathology , Obesity/complications , Disease Models, Animal , Signal Transduction
9.
Biomed Pharmacother ; 176: 116888, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38861859

ABSTRACT

OBJECTIVES: Co-agonists at the glucagon-like peptide-1 and glucagon receptors (GLP1R/GCGR) show promise as treatments for metabolic dysfunction-associated steatotic liver disease (MASLD). Although most co-agonists to date have been heavily GLP1R-biased, glucagon directly acts on the liver to reduce fat content. The aims of this study were to investigate a GCGR-biased co-agonist as treatment for hepatic steatosis in mice. METHODS: Mice with diet-induced obesity (DIO) were treated with Dicretin, a GLP1/GCGR co-agonist with high potency at the GCGR, Semaglutide (GLP1R monoagonist) or food restriction over 24 days, such that their weight loss was matched. Hepatic steatosis, glucose tolerance, hepatic transcriptomics, metabolomics and lipidomics at the end of the study were compared with Vehicle-treated mice. RESULTS: Dicretin lead to superior reduction of hepatic lipid content when compared to Semaglutide or equivalent weight loss by calorie restriction. Markers of glucose tolerance and insulin resistance improved in all treatment groups. Hepatic transcriptomic and metabolomic profiling demonstrated many changes that were unique to Dicretin-treated mice. These include some known targets of glucagon signaling and others with as yet unclear physiological significance. CONCLUSIONS: Our study supports the development of GCGR-biased GLP1/GCGR co-agonists for treatment of MASLD and related conditions.


Subject(s)
Fatty Liver , Glucagon-Like Peptide 1 , Mice, Inbred C57BL , Obesity , Receptors, Glucagon , Weight Loss , Animals , Obesity/drug therapy , Obesity/metabolism , Weight Loss/drug effects , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Male , Fatty Liver/drug therapy , Fatty Liver/metabolism , Mice , Glucagon-Like Peptide 1/metabolism , Diet, High-Fat/adverse effects , Liver/metabolism , Liver/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Insulin Resistance , Glucagon-Like Peptides/pharmacology
10.
Adv Sci (Weinh) ; 11(29): e2400819, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38837628

ABSTRACT

Glucagon receptor (GCGR) agonism offers potentially greater effects on the mitigation of hepatic steatosis. However, its underlying mechanism is not fully understood. Here, it screened tetraspanin CD9 might medicate hepatic effects of GCGR agonist. CD9 is decreased in the fatty livers of patients and upregulated upon GCGR activation. Deficiency of CD9 in the liver exacerbated diet-induced hepatic steatosis via complement factor D (CFD) regulated fatty acid metabolism. Specifically, CD9 modulated hepatic fatty acid synthesis and oxidation genes through regulating CFD expression via the ubiquitination-proteasomal degradation of FLI1. In addition, CD9 influenced body weight by modulating lipogenesis and thermogenesis of adipose tissue through CFD. Moreover, CD9 reinforcement in the liver alleviated hepatic steatosis, and blockage of CD9 abolished the remission of hepatic steatosis induced by cotadutide treatment. Thus, CD9 medicates the hepatic beneficial effects of GCGR signaling, and may server as a promising therapeutic target for hepatic steatosis.


Subject(s)
Fatty Liver , Tetraspanin 29 , Tetraspanin 29/metabolism , Tetraspanin 29/genetics , Animals , Mice , Humans , Fatty Liver/metabolism , Fatty Liver/drug therapy , Disease Models, Animal , Male , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Receptors, Glucagon/genetics , Mice, Inbred C57BL , Liver/metabolism , Liver/drug effects , Signal Transduction/drug effects
11.
Nat Chem ; 16(9): 1436-1444, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38755312

ABSTRACT

Several peptide dual agonists of the human glucagon receptor (GCGR) and the glucagon-like peptide-1 receptor (GLP-1R) are in development for the treatment of type 2 diabetes, obesity and their associated complications. Candidates must have high potency at both receptors, but it is unclear whether the limited experimental data available can be used to train models that accurately predict the activity at both receptors of new peptide variants. Here we use peptide sequence data labelled with in vitro potency at human GCGR and GLP-1R to train several models, including a deep multi-task neural-network model using multiple loss optimization. Model-guided sequence optimization was used to design three groups of peptide variants, with distinct ranges of predicted dual activity. We found that three of the model-designed sequences are potent dual agonists with superior biological activity. With our designs we were able to achieve up to sevenfold potency improvement at both receptors simultaneously compared to the best dual-agonist in the training set.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Machine Learning , Receptors, Glucagon , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Drug Design , Peptides/chemistry , Peptides/pharmacology , Amino Acid Sequence , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/chemistry
12.
Diabetologia ; 67(8): 1602-1615, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38705923

ABSTRACT

AIMS/HYPOTHESES: Glucagon and glucagon-like peptide-1 (GLP-1) are derived from the same precursor; proglucagon, and dual agonists of their receptors are currently being explored for the treatment of obesity and metabolic dysfunction-associated steatotic liver disease (MASLD). Elevated levels of endogenous glucagon (hyperglucagonaemia) have been linked with hyperglycaemia in individuals with type 2 diabetes but are also observed in individuals with obesity and MASLD. GLP-1 levels have been reported to be largely unaffected or even reduced in similar conditions. We investigated potential determinants of plasma proglucagon and associations of glucagon receptor signalling with metabolic diseases based on data from the UK Biobank. METHODS: We used exome sequencing data from the UK Biobank for ~410,000 white participants to identify glucagon receptor variants and grouped them based on their known or predicted signalling. Data on plasma levels of proglucagon estimated using Olink technology were available for a subset of the cohort (~40,000). We determined associations of glucagon receptor variants and proglucagon with BMI, type 2 diabetes and liver fat (quantified by liver MRI) and performed survival analyses to investigate if elevated proglucagon predicts type 2 diabetes development. RESULTS: Obesity, MASLD and type 2 diabetes were associated with elevated plasma levels of proglucagon independently of each other. Baseline proglucagon levels were associated with the risk of type 2 diabetes development over a 14 year follow-up period (HR 1.13; 95% CI 1.09, 1.17; n=1562; p=1.3×10-12). This association was of the same magnitude across strata of BMI. Carriers of glucagon receptor variants with reduced cAMP signalling had elevated levels of proglucagon (ß 0.847; 95% CI 0.04, 1.66; n=17; p=0.04), and carriers of variants with a predicted frameshift mutation had higher levels of liver fat compared with the wild-type reference group (ß 0.504; 95% CI 0.03, 0.98; n=11; p=0.04). CONCLUSIONS/INTERPRETATION: Our findings support the suggestion that glucagon receptor signalling is involved in MASLD, that plasma levels of proglucagon are linked to the risk of type 2 diabetes development, and that proglucagon levels are influenced by genetic variation in the glucagon receptor, obesity, type 2 diabetes and MASLD. Determining the molecular signalling pathways downstream of glucagon receptor activation may guide the development of biased GLP-1/glucagon co-agonist with improved metabolic benefits. DATA AVAILABILITY: All coding is available through https://github.com/nicwin98/UK-Biobank-GCG.


Subject(s)
Biological Specimen Banks , Diabetes Mellitus, Type 2 , Obesity , Proglucagon , Receptors, Glucagon , Signal Transduction , Humans , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , United Kingdom , Female , Proglucagon/metabolism , Proglucagon/genetics , Male , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Middle Aged , Obesity/blood , Aged , Adult , Body Mass Index , Glucagon/blood , Glucagon-Like Peptide 1/blood , UK Biobank
13.
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
14.
Mol Metab ; 85: 101947, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38677509

ABSTRACT

OBJECTIVE: Type 2 diabetes (T2D) is characterised by the loss of first-phase insulin secretion. We studied mice with ß-cell selective loss of the glucagon receptor (Gcgrfl/fl X Ins-1Cre), to investigate the role of intra-islet glucagon receptor (GCGR) signalling on pan-islet [Ca2+]I activity and insulin secretion. METHODS: Metabolic profiling was conducted on Gcgrß-cell-/- and littermate controls. Crossing with GCaMP6f (STOP flox) animals further allowed for ß-cell specific expression of a fluorescent calcium indicator. These islets were functionally imaged in vitro and in vivo. Wild-type mice were transplanted with islets expressing GCaMP6f in ß-cells into the anterior eye chamber and placed on a high fat diet. Part of the cohort received a glucagon analogue (GCG-analogue) for 40 days and the control group were fed to achieve weight matching. Calcium imaging was performed regularly during the development of hyperglycaemia and in response to GCG-analogue treatment. RESULTS: Gcgrß-cell-/- mice exhibited higher glucose levels following intraperitoneal glucose challenge (control 12.7 mmol/L ± 0.6 vs. Gcgrß-cell-/- 15.4 mmol/L ± 0.0 at 15 min, p = 0.002); fasting glycaemia was not different to controls. In vitro, Gcgrß-cell-/- islets showed profound loss of pan-islet [Ca2+]I waves in response to glucose which was only partially rescued in vivo. Diet induced obesity and hyperglycaemia also resulted in a loss of co-ordinated [Ca2+]I waves in transplanted islets. This was reversed with GCG-analogue treatment, independently of weight-loss (n = 8). CONCLUSION: These data provide novel evidence for the role of intra-islet GCGR signalling in sustaining synchronised [Ca2+]I waves and support a possible therapeutic role for glucagonergic agents to restore the insulin secretory capacity lost in T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon , Glucose , Homeostasis , Insulin Secretion , Insulin-Secreting Cells , Receptors, Glucagon , Signal Transduction , Animals , Glucagon/metabolism , Mice , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Receptors, Glucagon/metabolism , Receptors, Glucagon/genetics , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Male , Islets of Langerhans/metabolism , Mice, Inbred C57BL , Mice, Knockout , Diet, High-Fat , Blood Glucose/metabolism , Female
15.
Peptides ; 176: 171219, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38615717

ABSTRACT

People with obesity and type 2 diabetes have a high prevalence of metabolic-associated steatotic liver disease, hyperlipidemia and cardiovascular disease. Glucagon increases hepatic glucose production; it also decreases hepatic fat accumulation, improves lipidemia and increases energy expenditure. Pharmaceutical strategies to antagonize the glucagon receptor improve glycemic outcomes in people with diabetes and obesity, but they increase hepatic steatosis and worsen dyslipidemia. Co-agonism of the glucagon and glucagon-like peptide-1 (GLP-1) receptors has emerged as a promising strategy to improve glycemia in people with diabetes and obesity. Addition of glucagon receptor agonism enhances weight loss, reduces liver fat and ameliorates dyslipidemia. Prior to clinical use, however, further studies are needed to investigate the safety and efficacy of glucagon and GLP-1 receptor co-agonists in people with diabetes and obesity and related conditions, with specific concerns regarding a higher prevalence of gastrointestinal side effects, loss of muscle mass and increases in heart rate. Furthermore, co-agonists with differing ratios of glucagon:GLP-1 receptor activity vary in their clinical effect; the optimum balance is yet to be identified.


Subject(s)
Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Glucagon , Obesity , Humans , Obesity/drug therapy , Obesity/metabolism , Glucagon/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hypoglycemic Agents/therapeutic use , Hypoglycemic Agents/pharmacology , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism
16.
J Endocrinol ; 261(3)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38614123

ABSTRACT

The glucagon receptor family are typical class B1 G protein-coupled receptors (GPCRs) with important roles in metabolism, including the control of pancreas, brain, and liver function. As proteins with seven transmembrane domains, GPCRs are intimately in contact with lipid bilayers and therefore can be putatively regulated by interactions with their lipidic components, including cholesterol, sphingolipids, and other lipid species. Additionally, these receptors, as well as the agonists they bind to, can undergo lipid modifications, which can influence their binding capacity and/or elicit modified or biased signalling profiles. While the effect of lipids, and in particular cholesterol, has been widely studied for other GPCR classes, information about their role in regulating the glucagon receptor family is only beginning to emerge. Here we summarise our current knowledge on the effects of cholesterol modulation of glucagon receptor family signalling and trafficking profiles, as well as existing evidence for specific lipid-receptor binding and indirect effects of lipids via lipid modification of cognate agonists. Finally, we discuss the different methodologies that can be employed to study lipid-receptor interactions and summarise the importance of this area of investigation to increase our understanding of the biology of this family of metabolically relevant receptors.


Subject(s)
Cholesterol , Receptors, Glucagon , Signal Transduction , Humans , Receptors, Glucagon/metabolism , Animals , Cholesterol/metabolism , Signal Transduction/physiology , Lipid Metabolism/physiology
17.
Peptides ; 176: 171213, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38604379

ABSTRACT

Glucagon is best known for its contribution to glucose regulation through activation of the glucagon receptor (GCGR), primarily located in the liver. However, glucagon's impact on other organs may also contribute to its potent effects in health and disease. Given that glucagon-based medicine is entering the arena of anti-obesity drugs, elucidating extrahepatic actions of glucagon are of increased importance. It has been reported that glucagon may stimulate secretion of arginine-vasopressin (AVP)/copeptin, growth hormone (GH) and adrenocorticotrophic hormone (ACTH) from the pituitary gland. Nevertheless, the mechanisms and whether GCGR is present in human pituitary are unknown. In this study we found that intravenous administration of 0.2 mg glucagon to 14 healthy subjects was not associated with increases in plasma concentrations of copeptin, GH, ACTH or cortisol over a 120-min period. GCGR immunoreactivity was present in the anterior pituitary but not in cells containing GH or ACTH. Collectively, glucagon may not directly stimulate secretion of GH, ACTH or AVP/copeptin in humans but may instead be involved in yet unidentified pituitary functions.


Subject(s)
Adrenocorticotropic Hormone , Glucagon , Glycopeptides , Humans , Glycopeptides/metabolism , Glucagon/metabolism , Glucagon/blood , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Male , Adult , Female , Pituitary Gland/metabolism , Pituitary Gland/drug effects , Hydrocortisone/blood , Receptors, Glucagon/metabolism , Human Growth Hormone/metabolism , Growth Hormone/metabolism , Growth Hormone/blood , Middle Aged
18.
Am J Physiol Endocrinol Metab ; 326(6): E747-E766, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38477666

ABSTRACT

Chronic kidney disease is a debilitating condition associated with significant morbidity and mortality. In recent years, the kidney effects of incretin-based therapies, particularly glucagon-like peptide-1 receptor agonists (GLP-1RAs), have garnered substantial interest in the management of type 2 diabetes and obesity. This review delves into the intricate interactions between the kidney, GLP-1RAs, and glucagon, shedding light on their mechanisms of action and potential kidney benefits. Both GLP-1 and glucagon, known for their opposing roles in regulating glucose homeostasis, improve systemic risk factors affecting the kidney, including adiposity, inflammation, oxidative stress, and endothelial function. Additionally, these hormones and their pharmaceutical mimetics may have a direct impact on the kidney. Clinical studies have provided evidence that incretins, including those incorporating glucagon receptor agonism, are likely to exhibit improved kidney outcomes. Although further research is necessary, receptor polypharmacology holds promise for preserving kidney function through eliciting vasodilatory effects, influencing volume and electrolyte handling, and improving systemic risk factors.


Subject(s)
Incretins , Renal Insufficiency, Chronic , Humans , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/physiopathology , Incretins/therapeutic use , Incretins/pharmacology , Animals , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Kidney/drug effects , Kidney/metabolism , Glucagon/metabolism
19.
Biomed Pharmacother ; 174: 116485, 2024 May.
Article in English | MEDLINE | ID: mdl-38518602

ABSTRACT

BACKGROUND: Glucagon-like peptide-1 (GLP-1)/glucagon (GCG) dual receptor agonists with different receptor selectivity are under investigation and have shown significant improvement in both weight loss and glycemic control, but the optimal potency ratio between the two receptors to balance efficacy and safety remains unclear. EXPERIMENTAL APPROACH: We designed and constructed several dual receptor agonists with different receptor potency ratios using Fc fusion protein technology. The long-term effects of the candidates on body weight and metabolic dysfunction-associated steatotic liver disease (MASLD) were evaluated in diet-induced obese (DIO) model mice, high-fat diet (HFD)-ob/ob mice and AMLN diet-induced MASLD mice. Repeat dose toxicity assays were performed to investigate the safety profile of the candidate (HEC-C070) in Sprague Dawley (SD) rats. KEY RESULTS: The high GCG receptor (GCGR) selectivity of HEC-C046 makes it more prominent than other compounds for weight loss and most MASLD parameters but may lead to safety concerns. The weight change of HEC-C052 with the lowest GCG agonism was inferior to that of selective GLP-1 receptor agonist (GLP-1RA) semaglutide in DIO model mice. The GLP-1R selectivity of HEC-C070 with moderate GCG agonism has a significant effect on weight loss and liver function in obese mice, and its lowest observed adverse effect level (LOAEL) was 30 nmol/kg in the repeat dose toxicity study. CONCLUSION: We compared the potential of the Fc fusion protein GLP-1/GCG dual receptor agonists with different receptor selectivity to provide the setting for future GLP-1/GCG dual receptor agonists to treat obesity and MASLD.


Subject(s)
Diet, High-Fat , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Immunoglobulin Fc Fragments , Obesity , Receptors, Glucagon , Recombinant Fusion Proteins , Animals , Humans , Mice , Rats , Diet, High-Fat/adverse effects , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptides/pharmacology , Immunoglobulin Fc Fragments/pharmacology , Mice, Inbred C57BL , Mice, Obese , Obesity/drug therapy , Obesity/metabolism , Rats, Sprague-Dawley , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Recombinant Fusion Proteins/pharmacology , Weight Loss/drug effects
20.
Br J Pharmacol ; 181(12): 1874-1885, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38403793

ABSTRACT

BACKGROUND AND PURPOSE: Cotadutide is a dual GLP-1 and glucagon receptor agonist with balanced agonistic activity at each receptor designed to harness the advantages on promoting liver health, weight loss and glycaemic control. We characterised the effects of cotadutide on glucose, insulin, GLP-1, GIP, and glucagon over time in a quantitative manner using our glucose dynamics systems model (4GI systems model), in combination with clinical data from a multiple ascending dose/Phase 2a (MAD/Ph2a) study in overweight and obese subjects with a history of Type 2 diabetes mellitus (NCT02548585). EXPERIMENTAL APPROACH: The cotadutide PK-4GI systems model was calibrated to clinical data by re-estimating only food related parameters. In vivo cotadutide efficacy was scaled based on in vitro potency. The model was used to explore the effect of weight loss on insulin sensitivity and predict the relative contribution of the GLP-1 and glucagon receptor agonistic effects on glucose. KEY RESULTS: Cotadutide MAD/Ph2a clinical endpoints were successfully predicted. The 4GI model captured a positive effect of weight loss on insulin sensitivity and showed that the stimulating effect of glucagon on glucose production counteracts the GLP-1 receptor-mediated decrease in glucose, resulting in a plateau for glucose decrease around a 200-µg cotadutide dose. CONCLUSION AND IMPLICATIONS: The 4GI quantitative systems pharmacology model was able to predict the clinical effects of cotadutide on glucose, insulin, GLP-1, glucagon and GIP given known in vitro potency. The analyses demonstrated that the quantitative systems pharmacology model, and its successive refinements, will be a valuable tool to support the clinical development of cotadutide and related compounds.


Subject(s)
Blood Glucose , Diabetes Mellitus, Type 2 , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents , Models, Biological , Receptors, Glucagon , Humans , Receptors, Glucagon/agonists , Receptors, Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hypoglycemic Agents/pharmacology , Blood Glucose/drug effects , Blood Glucose/metabolism , Male , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glycemic Control , Middle Aged , Female , Adult , Glucagon/pharmacology , Glucagon/metabolism , Insulin/metabolism , Insulin/pharmacology , Obesity/drug therapy , Obesity/metabolism , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/pharmacology , Dose-Response Relationship, Drug , Peptides
SELECTION OF CITATIONS
SEARCH DETAIL