Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.133
1.
Int J Mol Sci ; 25(7)2024 Mar 29.
Article En | MEDLINE | ID: mdl-38612640

Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are incretins that regulate postprandial glucose regulation, stimulating insulin secretion from pancreatic ß-cells in response to food ingestion. Modified GLP-1 receptor agonists (GLP-1RAs) are being administered for the treatment of obesity and type 2 diabetes mellitus (T2DM). Strongly related to those disorders, metabolic dysfunction-associated steatotic liver disease (MASLD), especially its aggressive form, defined as metabolic dysfunction-associated steatohepatitis (MASH), is a major healthcare burden associated with high morbidity and extrahepatic complications. GLP-1RAs have been explored in MASH patients with evident improvement in liver dysfunction enzymes, glycemic control, and weight loss. Importantly, the combination of GLP-1RAs with GIP and/or glucagon RAs may be even more effective via synergistic mechanisms in amelioration of metabolic, biochemical, and histological parameters of MASLD but also has a beneficial impact on MASLD-related complications. In this current review, we aim to provide an overview of incretins' physiology, action, and signaling. Furthermore, we provide insight into the key pathophysiological mechanisms through which they impact MASLD aspects, as well as we analyze clinical data from human interventional studies. Finally, we discuss the current challenges and future perspectives pertinent to this growing area of research and clinical medicine.


Diabetes Mellitus, Type 2 , Digestive System Diseases , Fatty Liver , Liver Diseases , Metabolic Diseases , Humans , Receptors, Glucagon , Glucagon-Like Peptide 1/therapeutic use , Incretins/therapeutic use , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/therapeutic use , Receptors, G-Protein-Coupled
2.
JAAPA ; 37(5): 12-14, 2024 May 01.
Article En | MEDLINE | ID: mdl-38662894

ABSTRACT: Glucagon-like peptide 1 agonists (GLP1s) and the novel glucose-dependent insulinotropic polypeptide/glucagon-like peptide 1 agonist are effective drugs for reducing A1C and weight in patients with type 2 diabetes. However, clinicians may find it difficult to discern which drug to prescribe in specific clinical scenarios. This article discusses evidence-based clinical use of these drugs.


Diabetes Mellitus, Type 2 , Glucagon-Like Peptide 1 , Hypoglycemic Agents , Weight Loss , Humans , Diabetes Mellitus, Type 2/drug therapy , Weight Loss/drug effects , Glucagon-Like Peptide 1/agonists , Hypoglycemic Agents/therapeutic use , Liraglutide/therapeutic use , Gastric Inhibitory Polypeptide/therapeutic use , Gastric Inhibitory Polypeptide/agonists , Exenatide/therapeutic use , Exenatide/administration & dosage , Peptides/therapeutic use , Glycated Hemoglobin , Glucagon-Like Peptide-1 Receptor/agonists
3.
J Endocrinol ; 261(3)2024 Jun 01.
Article En | MEDLINE | ID: mdl-38579777

Adipose tissue was once known as a reservoir for energy storage but is now considered a crucial organ for hormone and energy flux with important effects on health and disease. Glucose-dependent insulinotropic polypeptide (GIP) is an incretin hormone secreted from the small intestinal K cells, responsible for augmenting insulin release, and has gained attention for its independent and amicable effects with glucagon-like peptide 1 (GLP-1), another incretin hormone secreted from the small intestinal L cells. The GIP receptor (GIPR) is found in whole adipose tissue, whereas the GLP-1 receptor (GLP-1R) is not, and some studies suggest that GIPR action lowers body weight and plays a role in lipolysis, glucose/lipid uptake/disposal, adipose tissue blood flow, lipid oxidation, and free-fatty acid (FFA) re-esterification, which may or may not be influenced by other hormones such as insulin. This review summarizes the research on the effects of GIP in adipose tissue (distinct depots of white and brown) using cellular, rodent, and human models. In doing so, we explore the mechanisms of GIPR-based medications for treating metabolic disorders, such as type 2 diabetes and obesity, and how GIPR agonism and antagonism contribute to improvements in metabolic health outcomes, potentially through actions in adipose tissues.


Adipose Tissue , Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Humans , Gastric Inhibitory Polypeptide/metabolism , Animals , Adipose Tissue/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Glucose/metabolism , Lipolysis , Obesity/metabolism
4.
Discov Med ; 36(183): 655-665, 2024 Apr.
Article En | MEDLINE | ID: mdl-38665015

Incretin hormones, such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 and 2 (GLP-1, 2), belong to the group of gastrointestinal hormones. Their actions occur through interaction with GIP and GLP-1/2 receptors, which are present in various target tissues. Apart from their well-established roles in pancreatic function and insulin regulation, incretins elicit significant effects that extend beyond the pancreas. Specifically, these hormones stimulate osteoblast differentiation and inhibit osteoclast activity, thereby promoting bone anabolism. Moreover, they play a pivotal role in bone mineralization and overall bone quality and function, making them potentially therapeutic for managing bone health. Thus, this review provides a summary of the crucial involvement of incretins in bone metabolism, influencing both bone formation and resorption processes. While existing evidence is persuasive, further studies are necessary for a comprehensive understanding of the therapeutic potential of incretins in modifying bone health.


Bone Remodeling , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide 1 , Glucagon-Like Peptide 2 , Incretins , Humans , Bone Remodeling/drug effects , Gastric Inhibitory Polypeptide/metabolism , Incretins/therapeutic use , Incretins/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 2/metabolism , Animals , Bone and Bones/metabolism , Bone and Bones/drug effects , Pancreas/metabolism , Pancreas/drug effects , Pancreas/pathology
5.
Expert Opin Drug Discov ; 19(5): 511-522, 2024 May.
Article En | MEDLINE | ID: mdl-38654653

INTRODUCTION: Despite numerous antidiabetic medications available for the treatment of type 2 diabetes, a substantial percentage of patients fail to achieve optimal glycemic control. Furthermore, the escalating obesity pandemic underscores the urgent need for effective relevant pharmacotherapies. Tirzepatide, a novel dual GIP and GLP-1 receptor agonist, offers a promising therapeutic option. AREAS COVERED: This review describes the discovery and clinical development of tirzepatide. Based on data from pivotal in vivo and in vitro studies, the authors present the pharmacodynamic profile of tirzepatide. Furthermore, they summarize data from the clinical trial programs that assessed the efficacy and safety of tirzepatide for the treatment of type 2 diabetes or obesity in a broad spectrum of patients, and discuss its therapeutic potential. EXPERT OPINION: Tirzepatide effectively reduces glucose levels and body weight in patients with type 2 diabetes and/or obesity, with a generally safe profile. Based on data from phase 3 clinical trials, several agencies have approved its use for the treatment of type 2 diabetes and obesity. Clinicians should be aware of possible adverse events, mainly mild-to-moderate gastrointestinal side effects. Overall, tirzepatide represents a promising treatment option for the treatment of type 2 diabetes.


Diabetes Mellitus, Type 2 , Drug Development , Drug Discovery , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-1 Receptor , Glucagon-Like Peptide-2 Receptor , Hypoglycemic Agents , Obesity , Diabetes Mellitus, Type 2/drug therapy , Humans , Animals , Glucagon-Like Peptide-1 Receptor/agonists , Obesity/drug therapy , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/administration & dosage , Blood Glucose/drug effects
6.
Sci Rep ; 14(1): 7557, 2024 03 30.
Article En | MEDLINE | ID: mdl-38555375

Although thorough chewing lowers postprandial glucose concentrations, research on the effectiveness of chewing vegetables in different forms on postprandial glucose metabolism remains limited. This study examined the effects of vegetables consumed in solid versus puree forms on postprandial glucose metabolism. Nineteen healthy young men completed two 180-min trials on separate days in a random order: the chewing trial involved the consumption of shredded cabbage with chewing and the non-chewing trial involved the consumption of pureed cabbage without chewing. Energy jelly was consumed immediately after the consumption of shredded or puree cabbage. Blood samples were collected at 0, 30, 45, 60, 90, 120 and 180 min. Circulating concentrations of glucose, insulin, total glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) concentrations were measured from the plasma. Although plasma glucose concentrations did not differ between the trials, the plasma insulin and GIP incremental area under the curve values were higher in the chewing than in the non-chewing trial. Postprandial total GLP-1 concentrations were higher in the chewing than in the non-chewing trial at 45, 60 and 90 min. This study demonstrates that consuming shredded cabbage while chewing enhances postprandial incretin secretion but has no effect on postprandial glucose concentration.Trial registration: Clinical trial registration ID.: UMIN000052662, registered 31 October 2023.


Blood Glucose , Vegetables , Male , Humans , Vegetables/metabolism , Blood Glucose/metabolism , Mastication , Glucose/metabolism , Insulin/metabolism , Glucagon-Like Peptide 1/metabolism , Gastric Inhibitory Polypeptide , Postprandial Period
7.
Spectrochim Acta A Mol Biomol Spectrosc ; 313: 124159, 2024 May 15.
Article En | MEDLINE | ID: mdl-38508074

The present study is focused on the sensitive determination of newly FDA-approved glucagon-like-peptide agonists semaglutide (SEM) and tirzepatide (TIR). Direct, selective and label-free spectrofluorometric method was proposed and validated (according to ICH guidelines) for determination SEM and TIR in their pure form, newly approved pharmaceuticals and spiked human plasma. The developed method was based on measuring the native fluorescence of SEM and TIR in ethanol at 294.8 and 303 nm after being excited at 216 and 225 nm for SEM and TIR in order. The method sensibility allowed the quantification of both drugs in nano-scale up to 10 ng/mL. Several experimental variables including solvent type, surfactant, and pH were optimized after several attempts to get the best sensitivity for both drugs. The mean recovery percentage of SEM was compared and found in agreement with the reported method using student's t-test and the variance ratio F-test. Additionally, the greenness and whiteness profiles for this approach were evaluated using the GAPI, AGREE, and RGB algorithm; the positive results supported its use as great candidates for successful implementation in quality control labs and the pharmaceutical analysis companies.


Algorithms , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Glucagon-Like Peptides , Humans , Coloring Agents
8.
Cardiovasc Diabetol ; 23(1): 112, 2024 Mar 30.
Article En | MEDLINE | ID: mdl-38555463

BACKGROUND: Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are effective antidiabetic drugs with potential cardiovascular benefits. Despite their well-established role in reducing the risk of major adverse cardiovascular events (MACE), their impact on heart failure (HF) remains unclear. Therefore, our study examined the cardioprotective effects of tirzepatide (TZT), a novel glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) receptor agonist. METHODS: A three-steps approach was designed: (i) Meta-analysis investigation with the primary objective of assessing major adverse cardiovascular events (MACE) occurrence from major randomized clinical trials.; (ii) TZT effects on a human cardiac AC16 cell line exposed to normal (5 mM) and high (33 mM) glucose concentrations for 7 days. The gene expression and protein levels of primary markers related to cardiac fibrosis, hypertrophy, and calcium modulation were evaluated. (iii) In silico data from bioinformatic analyses for generating an interaction map that delineates the potential mechanism of action of TZT. RESULTS: Meta-analysis showed a reduced risk for MACE events by TZT therapy (HR was 0.59 (95% CI 0.40-0.79, Heterogeneity: r2 = 0.01, I2 = 23.45%, H2 = 1.31). In the human AC16 cardiac cell line treatment with 100 nM TZT contrasted high glucose (HG) levels increase in the expression of markers associated with fibrosis, hypertrophy, and cell death (p < 0.05 for all investigated markers). Bioinformatics analysis confirmed the interaction between the analyzed markers and the associated pathways found in AC16 cells by which TZT affects apoptosis, fibrosis, and contractility, thus reducing the risk of heart failure. CONCLUSION: Our findings indicate that TZT has beneficial effects on cardiac cells by positively modulating cardiomyocyte death, fibrosis, and hypertrophy in the presence of high glucose concentrations. This suggests that TZT may reduce the risk of diabetes-related cardiac damage, highlighting its potential as a therapeutic option for heart failure management clinical trials. Our study strongly supports the rationale behind the clinical trials currently underway, the results of which will be further investigated to gain insights into the cardiovascular safety and efficacy of TZT.


Diabetes Mellitus, Type 2 , Diabetes Mellitus , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Heart Failure , Humans , Heart Failure/prevention & control , Diabetes Mellitus/diagnosis , Diabetes Mellitus/drug therapy , Hypertrophy , Hypoglycemic Agents/pharmacology , Myocytes, Cardiac , Fibrosis , Glucose , Glucagon-Like Peptide-1 Receptor
9.
BMC Endocr Disord ; 24(1): 38, 2024 Mar 14.
Article En | MEDLINE | ID: mdl-38481208

BACKGROUND: Glucagon is secreted from pancreatic alpha cells in response to low blood glucose and increases hepatic glucose production. Furthermore, glucagon enhances hepatic protein and lipid metabolism during a mixed meal. Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are secreted from gut endocrine cells during meals and control glucose homeostasis by potentiating insulin secretion and inhibiting food intake. Both glucose homeostasis and food intake have been reported to be affected by circadian rhythms and vice versa. In this study, we investigated whether the secretion of glucagon, GLP-1 and GIP was affected by circadian rhythms. METHODS: A total of 24 healthy men with regular sleep schedules were examined for 24 h at the hospital ward with 15 h of wakefulness and 9 h of sleep. Food intake was standardized, and blood samples were obtained every third hour. Plasma concentrations of glucagon, GLP-1 and GIP were measured, and data were analyzed by rhythmometric statistical methods. Available data on plasma glucose and plasma C-peptide were also included. RESULTS: Plasma concentrations of glucagon, GLP-1, GIP, C-peptide and glucose fluctuated with a diurnal 24-h rhythm, with the highest levels during the day and the lowest levels during the night: glucagon (p < 0.0001, peak time 18:26 h), GLP-1 (p < 0.0001, peak time 17:28 h), GIP (p < 0.0001, peak time 18:01 h), C-peptide (p < 0.0001, peak time 17.59 h), and glucose (p < 0.0001, peak time 23:26 h). As expected, we found significant correlations between plasma concentrations of C-peptide and GLP-1 and GIP but did not find correlations between glucose concentrations and concentrations of glucagon, GLP-1 and GIP. CONCLUSIONS: Our results demonstrate that under meal conditions that are similar to that of many free-living individuals, plasma concentrations of glucagon, GLP-1 and GIP were observed to be higher during daytime and evening than overnight. These findings underpin disturbed circadian rhythm as a potential risk factor for diabetes and obesity. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT06166368. Registered 12 December 2023.


Glucagon-Like Peptide 1 , Glucagon , Male , Humans , Glucagon/metabolism , Insulin , C-Peptide , Gastric Inhibitory Polypeptide , Blood Glucose/metabolism , Glucose/pharmacology , Circadian Rhythm
10.
Obes Rev ; 25(5): e13717, 2024 May.
Article En | MEDLINE | ID: mdl-38463003

Potent incretin-based therapy shows promise for the treatment of obesity along with reduced incidence of cardiovascular events in patients with preexisting cardiovascular disease and obesity. This study assessed the efficacy and safety of the incretin-based obesity treatments, once-weekly subcutaneous semaglutide 2.4 mg and tirzepatide 10 or 15 mg, in people with obesity without diabetes. Of the 744 records identified, seven randomized controlled trials (n = 5140) were included. Five studies (n = 3288) investigated semaglutide and two studies (n = 1852) investigated tirzepatide. The treatment effect, shown as placebo-subtracted difference, on body weight was -15.0% (95% CI, -17.8 to -12.2) with -12.9% (95% CI, -14.7 to -11.1) for semaglutide and -19.2% (95% CI, -22.2 to -16.2) for tirzepatide. The treatment effect on waist circumference was -11.4 cm (95% CI, -13.7 to -9.2) with -9.7 cm (95% CI, -10.8 to -8.5) for semaglutide and -14.6 cm (95% CI, -15.8 to -13.4) for tirzepatide. The adverse events related to semaglutide and tirzepatide were primarily of mild-to-moderate severity and mostly gastrointestinal, which was more frequent during the dose-titration period and leveled off during the treatment period. This emphasizes that once-weekly subcutaneous semaglutide 2.4 mg and tirzepatide 10 or 15 mg induce large reductions in body weight and waist circumference and are generally well-tolerated.


Diabetes Mellitus, Type 2 , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Glucagon-Like Peptides , Incretins , Humans , Incretins/therapeutic use , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/epidemiology , Waist Circumference , Body Weight , Obesity/drug therapy , Obesity/chemically induced , Glucagon-Like Peptide-1 Receptor
11.
Mol Cell Endocrinol ; 587: 112201, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38494045

The gut plays a crucial role in metabolism by regulating the passage of nutrients, water and microbial-derived substances to the portal circulation. Additionally, it produces incretins, such as glucose-insulinotropic releasing peptide (GIP) and glucagon-like derived peptide 1 (GLP1, encoded by gcg gene) in response to nutrient uptake. We aimed to investigate whether offspring from overweight rats develop anomalies in the barrier function and incretin transcription. We observed pro-inflammatory related changes along with a reduction in Claudin-3 levels resulting in increased gut-permeability in fetuses and offspring from overweight rats. Importantly, we found decreased gip mRNA levels in both fetuses and offspring from overweight rats. Differently, gcg mRNA levels were upregulated in fetuses, downregulated in female offspring and unchanged in male offspring from overweight rats. When cultured with high glucose, intestinal explants showed an increase in gip and gcg mRNA levels in control offspring. In contrast, offspring from overweight rats did not exhibit any response in gip mRNA levels. Additionally, while females showed no response, male offspring from overweight rats did exhibit an upregulation in gcg mRNA levels. Furthermore, female and male offspring from overweight rats showed sex-dependent anomalies when orally challenged with a glucose overload, returning to baseline glucose levels after 120 min. These results open new research questions about the role of the adverse maternal metabolic condition in the programming of impairments in glucose homeostasis, enteroendocrine function and gut barrier function in the offspring from overweight mothers and highlight the importance of a perinatal maternal healthy metabolism.


Gastric Inhibitory Polypeptide , Overweight , Rats , Male , Female , Animals , Overweight/metabolism , Gastric Inhibitory Polypeptide/metabolism , Incretins/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Peptides/metabolism , Homeostasis , RNA, Messenger/genetics
12.
Rev Med Suisse ; 20(866): 570-574, 2024 Mar 20.
Article Fr | MEDLINE | ID: mdl-38506456

The management of obesity is changing dramatically with the emergence of new drug treatments. Glucagon-like peptide-1 (GLP-1) receptor agonists are approved for this indication in Switzerland, and approval is currently being sought for a GLP-1 and glucose-dependent insulinotropic polypetide (GIP) co-agonist. Reimbursement conditions are restrictive, and patients are given only one opportunity to achieve the weight loss required for continued reimbursement. The popularity of these treatments has led to worldwide stock-outs for several months now, and it is essential that prescribers respect the indications so as not to prejudice obese patients. This article provides a review of the treatments available and the conditions under which they are reimbursed, as well as those that should be reimbursed soon.


La prise en charge de l'obésité est profondément modifiée par l'essor de nouveaux traitements médicamenteux. Les agonistes du récepteur du glucagon-like peptide-1 (GLP-1) sont approuvés dans cette indication en Suisse et une approbation est en cours d'analyse pour un coagoniste du GLP-1 et du glucose-dependent insulinotropic polypetide (GIP). Les conditions de remboursement sont cependant strictes et les patients ne bénéficient que d'une seule opportunité pour obtenir les pertes pondérales qui autorisent la poursuite du remboursement. La popularité de ces traitements occasionne des ruptures de stocks depuis plusieurs mois au niveau mondial et il est essentiel que les prescripteurs respectent les indications afin de ne pas prétériter les patients en situation d'obésité. Cet article propose un rappel des traitements disponibles et les conditions encadrant leur remboursement, ainsi que ceux qui le seront prochainement.


Blood Glucose , Diabetes Mellitus, Type 2 , Humans , Gastric Inhibitory Polypeptide , Obesity/drug therapy , Glucagon-Like Peptide 1 , Glucose , Glucagon-Like Peptide-1 Receptor/agonists
13.
J Med Chem ; 67(6): 4998-5010, 2024 Mar 28.
Article En | MEDLINE | ID: mdl-38458970

Glucose-dependent insulinotropic peptide (GIP) is a 42-amino acid peptide hormone that regulates postprandial glucose levels. GIP binds to its cognate receptor, GIPR, and mediates metabolic physiology by improved insulin sensitivity, ß-cell proliferation, increased energy consumption, and stimulated glucagon secretion. Dipeptidyl peptidase-4 (DPP4) catalyzes the rapid inactivation of GIP within 6 min in vivo. Here, we report a molecular platform for the design of GIP analogues that are refractory to DPP4 action and exhibit differential activation of the receptor, thus offering potentially hundreds of GIP-based compounds to fine-tune pharmacology. The lead compound from our studies, which harbored a combination of N-terminal alkylation and side-chain lipidation, was equipotent and retained full efficacy at GIPR as the native peptide, while being completely refractory toward DPP4, and was resistant to trypsin. The GIP analogue identified from these studies was further evaluated in vivo and is one of the longest-acting GIPR agonists to date.


Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/chemistry , Gastric Inhibitory Polypeptide/metabolism , Insulin/metabolism , Dipeptidyl Peptidase 4/metabolism , Peptide Hydrolases , Peptides , Endopeptidases , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/metabolism
14.
Obes Surg ; 34(4): 1324-1332, 2024 Apr.
Article En | MEDLINE | ID: mdl-38430320

BACKGROUND: Metabolic and bariatric surgery (MBS) is the most effective treatment for obesity and improvement of obesity-associated comorbidities. However, a proportion of these patients may suffer from weight recurrence and recurrence of obesity-associated comorbidities. METHOD: A retrospective cohort study of patients who underwent SG between January 2008 and August 2022 and sought treatment for weight recurrence with semaglutide or tirzepetide from January 2022 onwards. RESULT: A total of 115 patients were included, of which 70 had SG and treated for weight recurrence with semaglutide and 45 had SG and treated with tirzepatide. The mean age of patients was 38.8 (10.4) and 80.9% of patients were female. The mean pre-treatment weight and BMI was 94.0 (23.8) kg and 35.1 (6.0) kg/m2. Following treatment with semaglutide and tirzepatide, the mean post-treatment weight at 6 months was 81.0 (19.0) kg from 90.1 (19.6) kg and 87.6 (28.3) kg from 100.2 (28.5) kg respectively, corresponding to a clinically significant mean weight loss from baseline to 6 months of 10.3 (5.9)% (p < 0.05) and 15.5 (6.3)% (p < 0.05). Weight loss in tirzepatide patients was significantly greater than the semaglutide patients at 6 months (p < 0.02). There were no reported severe adverse events to the treatment. CONCLUSION: Short-term outcomes show that semaglutide and tirzepatide can be an effective treatment for managing weight recurrence after SG. Studies with longer follow-up are needed to determine the durability, as weight regain after discontinuation of the medication is highly likely, and the high cost of these medications can limit their use.


Gastric Inhibitory Polypeptide , Glucagon-Like Peptide-2 Receptor , Glucagon-Like Peptides , Obesity, Morbid , Humans , Female , Male , Obesity, Morbid/surgery , Retrospective Studies , Obesity/surgery , Treatment Outcome , Gastrectomy/adverse effects , Weight Loss
15.
Curr Opin Endocrinol Diabetes Obes ; 31(3): 115-121, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38511400

PURPOSE OF REVIEW: Various gut hormones interact with the brain through delicate communication, thereby influencing appetite and subsequent changes in body weight. This review summarizes the effects of gut hormones on appetite, with a focus on recent research. RECENT FINDINGS: Ghrelin is known as an orexigenic hormone, whereas glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), cholecystokinin (CCK), postprandial peptide YY (PYY), and oxyntomodulin (OXM) are known as anorexigenic hormones. Recent human studies have revealed that gut hormones act differently in various systems, including adipose tissue, beyond appetite and energy intake, and even involve in high-order thinking. Environmental factors including meal schedule, food contents and quality, type of exercise, and sleep deprivation also play a role in the influence of gut hormone on appetite, weight change, and obesity. Recently published studies have shown that retatrutide, a triple-agonist of GLP-1, GIP, and glucagon receptor, and orforglipron, a GLP-1 receptor partial agonist, are effective in weight loss and improving various metabolic parameters associated with obesity. SUMMARY: Various gut hormones influence appetite, and several drugs targeting these receptors have been reported to exert positive effects on weight loss in humans. Given that diverse dietary and environmental factors affect the actions of gut hormones and appetite, there is a need for integrated and largescale long-term studies in this field.


Appetite Regulation , Gastrointestinal Hormones , Obesity , Humans , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/physiology , Appetite Regulation/physiology , Obesity/metabolism , Obesity/physiopathology , Cholecystokinin/physiology , Cholecystokinin/metabolism , Gastric Inhibitory Polypeptide/physiology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/physiology , Peptide YY/metabolism , Peptide YY/physiology , Oxyntomodulin , Animals , Ghrelin/physiology , Ghrelin/metabolism , Appetite/physiology , Appetite/drug effects
16.
Peptides ; 176: 171186, 2024 Jun.
Article En | MEDLINE | ID: mdl-38490484

Circulating insulin levels are known to be increased in people with higher body mass index (BMI) due to effects of adiposity on insulin resistance, whilst gut hormones have a more complex relationship, with fasting peptideYY (PYY) reported to be inversely related to BMI. This study aimed to further explore fasting and post prandial pancreatic and gut hormone concentrations in plasma samples from obese and non-obese participants. Participants with healthy BMI (n=15), overweight BMI (n=29) and obesity (n=161) had samples taken fasting and 30 min post mixed liquid meal for analysis of glucagon-like peptide-1 (GLP-1), PYY, glucose-dependent insulinotropic polypeptide (GIP), insulin and glucagon. Data visualiation used linear discriminant analysis for dimensionality reduction, to visualise the data and assess scaling of each hormone. Fasting levels of insulin, GIP and PYY were shown to be key classifiers between the 3 groups on ANCOVA analysis, with an observation of increased GIP levels in overweight, but not obese participants. In non-obese subjects, fasting GIP, PYY and insulin correlated with BMI, whereas in subjects with obesity only the pancreatic hormones glucagon and insulin correlated with BMI. Concentrations of total GLP-1 in the fasting state correlated strongly with glucagon levels, highlighting potential assay cross-reactivities. The study, which included a relatively large number of subjects with severe obesity, supported previous evidence of BMI correlating negatively with fasting PYY and positively with fasting insulin. The observation of increased fasting GIP levels in overweight but not obese participants deserves further validation and mechanistic investigation.


Body Mass Index , Fasting , Gastric Inhibitory Polypeptide , Glucagon-Like Peptide 1 , Insulin , Obesity , Peptide YY , Humans , Obesity/blood , Male , Female , Adult , Fasting/blood , Peptide YY/blood , Middle Aged , Glucagon-Like Peptide 1/blood , Gastric Inhibitory Polypeptide/blood , Insulin/blood , Postprandial Period , Glucagon/blood , Gastrointestinal Hormones/blood
17.
Peptides ; 176: 171198, 2024 Jun.
Article En | MEDLINE | ID: mdl-38527521

In recent years, significant progress has been made to pharmacologically combat the obesity pandemic, particularly with regard to biochemically tailored drugs that simultaneously target the receptors for glucagon-like peptide-1 (GLP-1) and the glucose-dependent insulinotropic polypeptide (GIP). But while the pharmacological benefits of GLP-1 receptor (GLP-1R) agonism are widely acknowledged, the role of the GIP system in regulating systems metabolism remains controversial. When given in adjunct to GLP-1R agonism, both agonism and antagonism of the GIP receptor (GIPR) improves metabolic outcome in preclinical and clinical studies, and despite persistent concerns about its potential obesogenic nature, there is accumulating evidence indicating that GIP has beneficial metabolic effects via central GIPR agonism. Nonetheless, despite growing recognition of the GIP system as a valuable pharmacological target, there remains great uncertainty as to where and how GIP acts in the brain to regulate metabolism, and how GIPR agonism may differ from GIPR antagonism in control of energy metabolism. In this review we highlight current knowledge on the central action of GIP, and discuss open questions related to its multifaceted biology in the brain and the periphery.


Energy Metabolism , Gastric Inhibitory Polypeptide , Receptors, Gastrointestinal Hormone , Signal Transduction , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/agonists , Humans , Energy Metabolism/drug effects , Signal Transduction/drug effects , Gastric Inhibitory Polypeptide/metabolism , Animals , Obesity/metabolism , Obesity/drug therapy , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Brain/metabolism
18.
Peptides ; 176: 171200, 2024 Jun.
Article En | MEDLINE | ID: mdl-38555054

Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones produced by enteroendocrine cells in the small intestine. Despite being produced in the gut, the leveraging of their role in potentiating glucose-stimulated insulin secretion, also known as the incretin effect, has distracted from discernment of direct intestinal signaling circuits. Both preclinical and clinical evidence have highlighted a role for the incretins in inflammation. In this review, we highlight the discoveries of GLP-1 receptor (GLP-1R)+ natural (TCRαß and TCRγδ) and induced (TCRαß+CD4+ cells and TCRαß+CD8αß+) intraepithelial lymphocytes. Both endogenous signaling and pharmacological activation of GLP-1R impact local and systemic inflammation, the gut microbiota, whole-body metabolism, as well as the control of GLP-1 bioavailability. While GIPR signaling has been documented to impact hematopoiesis, the impact of these bone marrow-derived cells in gut immunology is not well understood. We uncover gaps in the literature of the evaluation of the impact of sex in these GLP-1R and GIP receptor (GIPR) signaling circuits and provide speculations of the maintenance roles these hormones play within the gut in the fasting-refeeding cycles. GLP-1R agonists and GLP-1R/GIPR agonists are widely used as treatments for diabetes and weight loss, respectively; however, their impact on gut homeostasis has not been fully explored. Advancing our understanding of the roles of GLP-1R and GIPR signaling within the gut at homeostasis as well as metabolic and inflammatory diseases may provide targets to improve disease management.


Glucagon-Like Peptide-1 Receptor , Inflammation , Receptors, Gastrointestinal Hormone , Humans , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Receptors, Gastrointestinal Hormone/metabolism , Inflammation/metabolism , Inflammation/immunology , Animals , Immunomodulation , Gastrointestinal Microbiome/immunology , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Signal Transduction
19.
Mol Metab ; 83: 101915, 2024 May.
Article En | MEDLINE | ID: mdl-38492844

OBJECTIVE: The glucose-dependent insulinotropic polypeptide (GIP) decreases body weight via central GIP receptor (GIPR) signaling, but the underlying mechanisms remain largely unknown. Here, we assessed whether GIP regulates body weight and glucose control via GIPR signaling in cells that express the leptin receptor (Lepr). METHODS: Hypothalamic, hindbrain, and pancreatic co-expression of Gipr and Lepr was assessed using single cell RNAseq analysis. Mice with deletion of Gipr in Lepr cells were generated and metabolically characterized for alterations in diet-induced obesity (DIO), glucose control and leptin sensitivity. Long-acting single- and dual-agonists at GIPR and GLP-1R were further used to assess drug effects on energy and glucose metabolism in DIO wildtype (WT) and Lepr-Gipr knock-out (KO) mice. RESULTS: Gipr and Lepr show strong co-expression in the pancreas, but not in the hypothalamus and hindbrain. DIO Lepr-Gipr KO mice are indistinguishable from WT controls related to body weight, food intake and diet-induced leptin resistance. Acyl-GIP and the GIPR:GLP-1R co-agonist MAR709 remain fully efficacious to decrease body weight and food intake in DIO Lepr-Gipr KO mice. Consistent with the demonstration that Gipr and Lepr highly co-localize in the endocrine pancreas, including the ß-cells, we find the superior glycemic effect of GIPR:GLP-1R co-agonism over single GLP-1R agonism to vanish in Lepr-Gipr KO mice. CONCLUSIONS: GIPR signaling in cells/neurons that express the leptin receptor is not implicated in the control of body weight or food intake, but is of crucial importance for the superior glycemic effects of GIPR:GLP-1R co-agonism relative to single GLP-1R agonism.


Body Weight , Eating , Gastric Inhibitory Polypeptide , Mice, Knockout , Obesity , Receptors, Gastrointestinal Hormone , Receptors, Leptin , Animals , Male , Mice , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Glucagon-Like Peptide-1 Receptor/genetics , Glucose/metabolism , Leptin/metabolism , Mice, Inbred C57BL , Obesity/metabolism , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Gastrointestinal Hormone/genetics , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Signal Transduction
20.
Diabetes Obes Metab ; 26(6): 2446-2455, 2024 Jun.
Article En | MEDLINE | ID: mdl-38528819

AIMS: To describe the overall fat distribution patterns independent of body mass index (BMI) in participants with type 2 diabetes (T2D) in the SURPASS-3 MRI substudy by comparison with sex- and BMI-matched virtual control groups (VCGs) derived from the UK Biobank imaging study at baseline and Week 52. METHODS: For each study participant at baseline and Week 52 (N = 296), a VCG of ≥150 participants with the same sex and similar BMI was identified from the UK Biobank imaging study (N = 40 172). Average visceral adipose tissue (VAT), abdominal subcutaneous adipose tissue (aSAT) and liver fat (LF) levels and the observed standard deviations (SDs; standardized normal z-scores: z-VAT, z-aSAT and z-LF) were calculated based on the matched VCGs. Differences in z-scores between baseline and Week 52 were calculated to describe potential shifts in fat distribution pattern independent of weight change. RESULTS: Baseline fat distribution patterns were similar across pooled tirzepatide (5, 10 and 15 mg) and insulin degludec (IDeg) arms. Compared with matched VCGs, SURPASS-3 participants had higher baseline VAT (mean [SD] z-VAT +0.42 [1.23]; p < 0.001) and LF (z-LF +1.24 [0.92]; p < 0.001) but similar aSAT (z-aSAT -0.13 [1.11]; p = 0.083). Tirzepatide-treated participants had significant decreases in z-VAT (-0.18 [0.58]; p < 0.001) and z-LF (-0.54 [0.84]; p < 0.001) but increased z-aSAT (+0.11 [0.50]; p = 0.012). Participants treated with IDeg had a significant change in z-LF only (-0.46 [0.90]; p = 0.001), while no significant changes were observed for z-VAT (+0.13 [0.52]; p = 0.096) and z-aSAT (+0.09 [0.61]; p = 0.303). CONCLUSION: In this exploratory analysis, treatment with tirzepatide in people with T2D resulted in a significant reduction of z-VAT and z-LF, while z-aSAT was increased from an initially negative value, suggesting a possible treatment-related shift towards a more balanced fat distribution pattern with prominent VAT and LF loss.


Body Fat Distribution , Diabetes Mellitus, Type 2 , Humans , Diabetes Mellitus, Type 2/drug therapy , Male , Female , Middle Aged , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/diagnostic imaging , Aged , Hypoglycemic Agents/therapeutic use , Body Mass Index , Magnetic Resonance Imaging , Glucagon-Like Peptide-2 Receptor , Gastric Inhibitory Polypeptide
...