Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 39
Filtrer
1.
Am J Clin Nutr ; 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38996913

RÉSUMÉ

BACKGROUND: In humans, intraduodenal infusion of L-tryptophan (Trp) increases plasma concentrations of gastrointestinal hormones and stimulates pyloric pressures, both key determinants of gastric emptying and associated with potent suppression of energy intake. The stimulation of gastrointestinal hormones by Trp has been shown, in preclinical studies, to be enhanced by extracellular calcium and mediated in part by the calcium-sensing receptor. OBJECTIVE: To determine whether intraduodenal calcium can enhance the effects of Trp to stimulate gastrointestinal hormones and pyloric pressures, and if so, whether it is associated with greater suppression of energy intake, in healthy males. METHODS: Fifteen males with normal weight (mean±SD; age: 26±7 years; body mass index: 22±2 kg/m2), received on three separate occasions, 150-min intraduodenal infusions of 0, 500 or 1000 mg calcium (Ca), each combined with Trp (load: 0.1 kcal/min, with submaximal energy intake-suppressant effects) from t=75-150 min, in a randomized, double-blind, cross-over study. Plasma concentrations of GI hormones (gastrin, cholecystokinin, glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), peptide tyrosine-tyrosine (PYY)), and Trp, and antropyloroduodenal pressures were measured throughout. Immediately post-infusions (t=150-180 min), energy intake at a standardized buffet-style meal was quantified. RESULTS: In response to calcium alone, both 500-mg and 1000-mg doses stimulated PYY, while only the 1000-mg dose stimulated GLP-1 and pyloric pressures (all P<0.05). The 1000-mg dose also enhanced the effects of Trp to stimulate cholecystokinin and GLP-1, and both doses stimulated PYY, but, surprisingly, reduced the stimulation of GIP (all P<0.05). Both doses substantially and dose-dependently enhanced the effects of Trp to suppress energy intake (kcal; Ca-0+Trp: 1108±70, Ca-500+Trp: 961±90, Ca-1000+Trp: 922±96; P<0.05). CONCLUSIONS: Intraduodenal administration of calcium enhances the effect of Trp to stimulate plasma cholecystokinin, GLP-1 and PYY, and suppress energy intake, in health. These findings have potential implications for novel nutrient-based approaches to energy intake regulation in obesity. CLINICAL TRIAL REGISTRY: The trial was registered with the Australian New Zealand Clinical Trial Registry (www.anzctr.org.au; trial number: ACTRN12620001294943).

2.
Cardiovasc Res ; 2024 Jun 04.
Article de Anglais | MEDLINE | ID: mdl-38832935

RÉSUMÉ

AIMS: Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) are increasingly used to treat type 2 diabetes and obesity. Albeit cardiovascular outcomes generally improve, treatment with GLP-1 RAs is associated with increased heart rate, the mechanism of which is unclear. METHODS AND RESULTS: We employed a large animal model, the female landrace pig, and used multiple in-vivo and ex-vivo approaches including pharmacological challenges, electrophysiology and high-resolution mass spectrometry to explore how GLP-1 elicits an increase in heart rate. In anaesthetized pigs, neither cervical vagotomy, adrenergic blockers (alpha, beta or combined alpha-beta blockade), ganglionic blockade (hexamethonium) nor inhibition of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels (ivabradine) abolished the marked chronotropic effect of GLP-1. GLP-1 administration to isolated perfused pig hearts also increased heart rate, which was abolished by GLP-1 receptor blockade. Electrophysiological characterization of GLP-1 effects in vivo and in isolated perfused hearts localized electrical modulation to the atria and conduction system. In isolated sinus nodes, GLP-1 administration shortened action potential cycle length of pacemaker cells and shifted the site of earliest activation. The effect was independent of HCN blockade. Collectively, these data support a direct effect of GLP-1 on GLP-1 receptors within the heart. Consistently, single nucleus RNA sequencing (snRNAseq) showed GLP-1 receptor expression in porcine pacemaker cells. Quantitative phosphoproteomics analyses of sinus node samples revealed that GLP-1 administration leads to phosphorylation changes of calcium cycling proteins of the sarcoplasmic reticulum, known to regulate heart rate. CONCLUSION: GLP-1 has direct chronotropic effects on the heart mediated by GLP-1 receptors in pacemaker cells of the sinus node, inducing changes in action potential morphology and the leading pacemaker site through a calcium signaling response characterized by PKA-dependent phosphorylation of Ca2+ cycling proteins involved in pace making. Targeting the pacemaker calcium clock may be a strategy to lower heart rate in GLP-1 RA recipients.

3.
Diabet Med ; 41(6): e15320, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38551152

RÉSUMÉ

INTRODUCTION: Post-bariatric hypoglycaemia (PBH) is a rare yet disabling clinical condition, mostly reported after Roux-en-Y gastric bypass (RYGB) surgery. RYGB is one of the most widely used and effective bariatric procedures. The pathophysiology of PBH remains unclear, and treatment options are limited in effectiveness and/or carry significant side effects. Acarbose slows carbohydrates digestion and absorption and is generally considered first-line pharmacological treatment for PBH but its gastrointestinal side effects limit patient compliance. Canagliflozin inhibits intestinal and renal sodium-dependent glucose absorption and reduces postprandial excursions of glucose, insulin and incretins after RYGB - effects that could be beneficial in ameliorating PBH. AIMS: The trial aims to investigate how blood glucose levels are affected during daily living in subjects with PBH during treatment with canagliflozin or acarbose compared with placebo, and to study the meal-induced entero-endocrine mechanisms implied in the treatment responses. METHODS: In a double-blinded, randomized, crossover clinical trial, HypoBar I will investigate the effectiveness in reducing the risk of PBH, safety, ambulatory glucose profile and entero-endocrine responses when PBH is treated with canagliflozin 300 mg twice daily during a 4-week intervention period, compared with acarbose 50 mg thrice daily or placebo. ETHICS AND DISSEMINATION: HypoBar I is approved by the Local regulatory entities. Results will be published in peer-reviewed journals. CONCLUSION: If effective, well-tolerated and safe, canagliflozin could be a novel treatment for people with PBH. HypoBar I might also unravel new mechanisms underlying PBH, potentially identifying new treatment targets. TRIAL REGISTRATION: EudraCT number 2022-000157-87.


Sujet(s)
Acarbose , Canagliflozine , Hypoglycémie , Adulte , Femelle , Humains , Mâle , Adulte d'âge moyen , Jeune adulte , Acarbose/usage thérapeutique , Glycémie/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Canagliflozine/usage thérapeutique , Études croisées , Méthode en double aveugle , Dérivation gastrique/effets indésirables , Hypoglycémie/prévention et contrôle , Hypoglycémie/induit chimiquement , Hypoglycémiants/usage thérapeutique , Complications postopératoires/traitement médicamenteux , Complications postopératoires/prévention et contrôle , Essais contrôlés randomisés comme sujet , Inhibiteurs du cotransporteur sodium-glucose de type 2/usage thérapeutique
4.
Diabetes Obes Metab ; 25(1): 98-109, 2023 01.
Article de Anglais | MEDLINE | ID: mdl-36054143

RÉSUMÉ

AIM: The voltage-gated potassium channel Kv 11.1 is important for repolarizing the membrane potential in excitable cells such as myocytes, pancreatic α- and ß-cells. Moxifloxacin blocks the Kv 11.1 channel and increases the risk of hypoglycaemia in patients with diabetes. We investigated glucose regulation and secretion of glucoregulatory hormones in young people with and without moxifloxacin, a drug known to block the Kv 11.1 channel. MATERIALS AND METHODS: The effect of moxifloxacin (800 mg/day for 4 days) or placebo on glucose regulation was assessed in a randomized, double-blind, crossover study of young men and women (age 20-40 years and body mass index 18.5-27.5 kg/m2 ) without chronic disease, using 6-h oral glucose tolerance tests and continuous glucose monitoring. RESULTS: Thirty-eight participants completed the study. Moxifloxacin prolonged the QTcF interval and increased heart rate. Hypoglycaemia was more frequently observed with moxifloxacin, both during the 8 days of continuous glucose monitoring and during the oral glucose tolerance tests. Hypoglycaemia questionnaire scores were higher after intake of moxifloxacin. Moxifloxacin reduced the early plasma-glucose response (AUC0-30 min ) by 7% (95% CI: -9% to -4%, p < .01), and overall insulin response (AUC0-360 min ) decreased by 18% (95% CI: -24% to -11%, p < .01) and plasma glucagon increased by 17% (95% CI: 4%-33%, p = .03). Insulin sensitivity calculated as the Matsuda index increased by 11%, and MISI, an index of muscle insulin sensitivity, increased by 34%. CONCLUSIONS: In young men and women, moxifloxacin, a drug known to block the Kv 11.1 channel, increased QT interval, decreased glucose levels and was associated with increased muscle insulin sensitivity and more frequent episodes of hypoglycaemia.


Sujet(s)
Fluoroquinolones , Insulinorésistance , Humains , Femelle , Adolescent , Jeune adulte , Adulte , Moxifloxacine/effets indésirables , Fluoroquinolones/effets indésirables , Études croisées , Autosurveillance glycémique , Glycémie
5.
Pharmacol Res ; 176: 106058, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34995796

RÉSUMÉ

The intestinal hormones glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-2 (GLP-2) are key regulators of postprandial bone turnover in humans. We hypothesized that GIP and GLP-2 co-administration would provide stronger effect on bone turnover than administration of the hormones separately, and tested this using subcutaneous injections of GIP and GLP-2 alone or in combination in humans. Guided by these findings, we designed series of GIPR-GLP-2R co-agonists as template for new osteoporosis treatment. The clinical experiment was a randomized cross-over design including 10 healthy men administered subcutaneous injections of GIP and GLP-2 alone or in combination. The GIPR-GLP-2R co-agonists were characterized in terms of binding and activation profiles on human and rodent GIP and GLP-2 receptors, and their pharmacokinetic (PK) profiles were improved by dipeptidyl peptidase-4 protection and site-directed lipidation. Co-administration of GIP and GLP-2 in humans resulted in an additive reduction in bone resorption superior to each hormone individually. The GIPR-GLP-2R co-agonists, designed by combining regions of importance for cognate receptor activation, obtained similar efficacies as the two native hormones and nanomolar potencies on both human receptors. The PK-improved co-agonists maintained receptor activity along with their prolonged half-lives. Finally, we found that the GIPR-GLP-2R co-agonists optimized toward the human receptors for bone remodeling are not feasible for use in rodent models. The successful development of potent and efficacious GIPR-GLP-2R co-agonists, combined with the improved effect on bone metabolism in humans by co-administration, support these co-agonists as a future osteoporosis treatment.


Sujet(s)
Remodelage osseux/effets des médicaments et des substances chimiques , Peptide gastrointestinal/pharmacologie , Glucagon-like peptide 2/pharmacologie , Récepteur du peptide-2 similaire au glucagon/agonistes , Récepteur hormone gastrointestinale/agonistes , Adulte , Animaux , Cellules COS , Chlorocebus aethiops , Études croisées , Femelle , Peptide gastrointestinal/sang , Peptide gastrointestinal/pharmacocinétique , Glucagon-like peptide 2/sang , Glucagon-like peptide 2/pharmacocinétique , Récepteur du peptide-2 similaire au glucagon/génétique , Humains , Mâle , Souris de lignée C57BL , Ostéoporose/traitement médicamenteux , Récepteur hormone gastrointestinale/génétique , Méthode en simple aveugle , Jeune adulte
6.
Neurogastroenterol Motil ; 34(1): e14210, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34378827

RÉSUMÉ

OBJECTIVE: Neurotensin (NT) is released from enteroendocrine cells and lowers food intake in rodents. We evaluated postprandial NT secretion in humans after surgeries associated with accelerated small intestinal nutrient delivery, and after Roux-en-Y gastric bypass (RYGB) when glucagon-like peptide-1 (GLP-1) signalling and dipeptidyl peptidase 4 (DPP-4) were inhibited, and during pharmacological treatments influencing entero-pancreatic functions. METHODS: We measured NT concentrations in plasma from meal studies: (I) after truncal vagotomy with pyloroplasty (TVP), cardia resection +TVP (CTVP), and matched controls (n = 10); (II) after RYGB, sleeve gastrectomy (SG), and in matched controls (n = 12); (III) after RYGB (n = 11) with antagonism of GLP-1 signalling using exendin(9-39) and DPP-4 inhibition using sitagliptin; (IV) after RYGB (n = 11) during a run-in period and subsequent treatment with, sitagliptin, liraglutide (GLP-1 receptor agonist), verapamil (calcium antagonist), acarbose (alpha glucosidase inhibitor), and pasireotide (somatostatin analogue), respectively. RESULTS: (I) NT secretion was similar after TVP/CTVP (p = 0.9), but increased vs. controls (p < 0.0001). (II) NT secretion was increased after RYGB vs. SG and controls (p < 0.0001). NT responses were similar in SG and controls (p = 0.3), but early postprandial NT concentrations were higher after SG (p < 0.05). (III) Exendin (9-39) and sitagliptin did not change NT responses vs placebo (p > 0.2), but responses were lower during sitagliptin vs. exendin(9-39) (p = 0.03). (IV) Pasireotide suppressed NT secretion (p = 0.004). Sitagliptin tended to lower NT secretion (p = 0.08). Liraglutide, verapamil, and acarbose had no effect (p > 0.9). CONCLUSION: Neurotensin secretion is increased after surgeries associated with accelerated gastric emptying and lowered by pasireotide.


Sujet(s)
Gastrectomie , Dérivation gastrique , Neurotensine/sang , Obésité/chirurgie , Vagotomie tronculaire , Glycémie , Glucagon-like peptide 1/sang , Humains , Hypoglycémiants/pharmacologie , Hypoglycémiants/usage thérapeutique , Liraglutide/administration et posologie , Liraglutide/usage thérapeutique , Obésité/sang , Obésité/traitement médicamenteux , Période post-prandiale
7.
J Clin Endocrinol Metab ; 107(1): 77-87, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34508600

RÉSUMÉ

CONTEXT: The colon houses most of humans' gut microbiota, which ferments indigestible carbohydrates. The products of fermentation have been proposed to influence the secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) from the many endocrine cells in the colonic epithelium. However, little is known about the colonic contribution to fasting or postprandial plasma levels of L-cell products. OBJECTIVE: To determine the impact of colonic lactulose fermentation on gut peptide secretion and to evaluate whether colonic endocrine secretion contributes to gut hormone concentrations measurable in the fasting state. METHODS: Ten healthy young men were studied on 3 occasions after an overnight fast. On 2 study days, lactulose (20 g) was given orally and compared to water intake on a third study day. For 1 of the lactulose visits, participants underwent a full colonic evacuation. Over a 6-h study protocol, lactulose fermentation was assessed by measuring exhaled hydrogen, and gut peptide secretion, paracetamol, and short-chain fatty acid levels were measured in plasma. RESULTS: Colonic evacuation markedly reduced hydrogen exhalation after lactulose intake (P = 0.013). Our analysis suggests that the colon does not account for the measurable amounts of GLP-1 and PYY present in the circulation during fasting and that fermentation and peptide secretion are not acutely related. CONCLUSION: Whether colonic luminal contents affect colonic L-cell secretion sufficiently to influence circulating concentrations requires further investigation. Colonic evacuation markedly reduced lactulose fermentation, but hormone releases were unchanged in the present study.


Sujet(s)
Côlon/métabolisme , Microbiome gastro-intestinal/physiologie , Muqueuse intestinale/métabolisme , Lactulose/métabolisme , Administration par voie orale , Adulte , Côlon/microbiologie , Études croisées , Fermentation , Glucagon-like peptide 1/sang , Glucagon-like peptide 1/métabolisme , Volontaires sains , Humains , Muqueuse intestinale/microbiologie , Lactulose/administration et posologie , Mâle , Peptide YY/sang , Peptide YY/métabolisme , Jeune adulte
8.
J Clin Endocrinol Metab ; 107(1): e188-e204, 2022 01 01.
Article de Anglais | MEDLINE | ID: mdl-34479362

RÉSUMÉ

CONTEXT: Entero-pancreatic hormone secretion has been reported during the pre-absorptive cephalic and gastric meal phases, but never with a blood sampling frequency providing a temporal resolution that allows close scrutiny and correlations with gastric emptying and glucose absorption. OBJECTIVE: We hypothesized that entero-pancreatic hormone secretion after nutrient ingestion would be rapid and correlate with gastric emptying and glucose absorption. METHODS: During 2 visits in a clinical research facility, 10 healthy young men ingested a 75-g glucose drink (OG) and a liquid mixed meal (LMM) (t = 0-2 minutes) on separate days. Acetaminophen and 3-O-methyl-D-glucopyranose (3-OMG) were added to the drinks to evaluate gastric emptying and glucose absorption, respectively. Arterialized venous blood was sampled (t = -30, -20, -18, -16, -14, -12, -10, -8, -6, -4, -2, 0, 2, 4, 6, 8, 10, 12, 14, 16, 18, 20, 30 minutes). Plasma glucose-dependent insulinotropic polypeptide (GIP), glucagon-like peptide-1 (GLP-1), gastrin, cholecystokinin (CCK), glucagon, pancreatic polypeptide (PP), 3-OMG, and glucose were measured, as were serum insulin, C-peptide, and acetaminophen. RESULTS: Acetaminophen increased 8 minutes after OG (P < 0.001) and LMM (P < 0.05); 3-OMG, 8 minutes after LMM (P < 0.0001), 10 minutes after OG (P = 0.04); PP, 4 minutes after LMM (P < 0.03); gastrin, 6 minutes after LMM (P < 0.003) and OG (P < 0.003); CCK, 6 minutes after LMM (P = 0.0001); GIP, 8 minutes after OG (P < 0.05) and LMM (P < 0.03); glucose, 8 minutes after OG (P < 0.001); 12 minutes after LMM (P < 0.02); GLP-1, 12 minutes after OG (P < 0.01), 10 minutes after LMM (P < 0.01); insulin, 12 minutes after LMM (P = 0.02) and OG (P = 0.002); C-peptide, 12 minutes after OG (P = 0.002) and LMM (P = 0.04). CONCLUSION: Early postprandial hormone responses show characteristic differences with regard to timing and amplitude but also great individual differences. This should be considered when interpreting mean responses and designing study protocols.


Sujet(s)
Marqueurs biologiques/sang , Vidange gastrique , Glucose/métabolisme , Repas , Hormones pancréatiques/sang , Adulte , Peptide C/sang , Cholécystokinine/sang , Études de suivi , Glucagon/sang , Glucagon-like peptide 1/sang , Humains , Insuline/sang , Mâle , Pronostic , Études prospectives , Jeune adulte
9.
Scand J Clin Lab Invest ; 82(1): 75-83, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34935574

RÉSUMÉ

Glucagon is a key regulator of metabolism and is used in the diagnostic of neuroendocrine tumors. Accurate measurement of glucagon requires both extreme sensitivity and specificity since several peptides are derived from the same proglucagon precursor encoding part of the glucagon sequence and given that glucagon circulates in picomolar concentrations. A sandwich ELISA was recently developed and extensively evaluated; however, this method may not be accurate when measuring glucagon in patients with an enhanced production of proglucagon-derived peptides as seen after Roux-en-Y gastric bypass (RYGB). To overcome this, a modified version of the ELISA was developed. In this study, we evaluate an unmodified and a modified version of the ELISA in healthy individuals, individuals with obesity, and finally in two cohorts of patients following RYGB surgery using different nutrient stimuli to assess glucagon dynamics. Finally, in vitro spike-in recoveries using native glucagon and proglucagon-derived peptides were performed in buffer and in plasma. Our data support that both versions of the ELISA accurately capture endogenous and exogenous glucagon in healthy individuals and in individuals with obesity. However, the unmodified version of the assay may overestimate glucagon levels in patients following RYGB in line with minimal but consistent cross-reactivity to oxyntomodulin and glicentin that both are 50-fold increased after RYGB. Importantly, we did not find any changes between the two protocols at fasted conditions and therefore diagnostics of glucagonomas is not affected by the choice of assay procedure nor the surgical history of the patient (RYGB).


Sujet(s)
Dérivation gastrique , Glycémie/analyse , Test ELISA , Dérivation gastrique/méthodes , Glucagon/métabolisme , Humains , Obésité/chirurgie , Proglucagon
10.
Front Endocrinol (Lausanne) ; 12: 681116, 2021.
Article de Anglais | MEDLINE | ID: mdl-34084153

RÉSUMÉ

Background: Altered bile acid (BA) turnover has been suggested to be involved in the improved glucose regulation after Roux-en-Y gastric bypass (RYGB), possibly via stimulation of GLP-1 secretion. We investigated the role of exogenous as well as endogenous BAs for GLP-1 secretion after RYGB by administering chenodeoxycholic acid (CDCA) and the BA sequestrant colesevelam (COL) both in the presence and the absence of a meal stimulus. Methods: Two single-blinded randomized cross-over studies were performed. In study 1, eight RYGB operated participants ingested 200 ml water with 1) CDCA 1.25 g or 2) CDCA 1.25 g + colesevelam 3.75 g on separate days. In study 2, twelve RYGB participants ingested on separate days a mixed meal with addition of 1) CDCA 1.25 g, 2) COL 3.75 g or 3) COL 3.75 g × 2, or 4) no additions. Results: In study 1, oral intake of CDCA increased circulating BAs, GLP-1, C-peptide, glucagon, and neurotensin. Addition of colesevelam reduced all responses. In study 2, addition of CDCA enhanced meal-induced increases in plasma GLP-1, glucagon and FGF-19 and lowered plasma glucose and C-peptide concentrations, while adding colesevelam lowered circulating BAs but did not affect meal-induced changes in plasma glucose or measured gastrointestinal hormones. Conclusion: In RYGB-operated persons, exogenous CDCA enhanced meal-stimulated GLP-1 and glucagon secretion but not insulin secretion, while the BA sequestrant colesevelam decreased CDCA-stimulated GLP-1 secretion but did not affect meal-stimulated GLP-1, C-peptide or glucagon secretion, or glucose tolerance. These findings suggest a limited role for endogenous bile acids in the acute regulation of postprandial gut hormone secretion or glucose metabolism after RYGB.


Sujet(s)
Acides et sels biliaires/sang , Dérivation gastrique , Glucagon-like peptide 1/sang , Glucose/métabolisme , Obésité morbide/chirurgie , Adulte , Glycémie , Peptide C/sang , Chlorhydrate de colésévélam/usage thérapeutique , Femelle , Glucagon/sang , Humains , Mâle , Adulte d'âge moyen , Neurotensine/sang , Obésité morbide/sang , Obésité morbide/traitement médicamenteux , Période post-prandiale , Méthode en simple aveugle
11.
Bone ; 152: 116065, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34153529

RÉSUMÉ

BACKGROUND: Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-2 (GLP-2) are gut hormones secreted in response to food ingestion, and they have been suggested to regulate bone turnover. In humans, exogenous GIP and GLP-2 acutely inhibit bone resorption as measured by circulating levels of carboxy-terminal type 1 collagen crosslinks (CTX). OBJECTIVE: The objective was to study the individual and combined acute effects of GIP and GLP-2 on bone turnover in postmenopausal women during nighttime - a period of increased bone resorption. METHODS: Using a randomized, placebo-controlled, double-blinded, crossover design, each participant (n = 9) received on four separate study days: GIP, GLP-2, GIP + GLP-2, and placebo (saline) as subcutaneous injections at bedtime. Main outcomes were levels of CTX and procollagen type 1 N-terminal propeptide (P1NP). RESULTS: Compared with placebo, GIP and GLP-2 alone significantly inhibited bone resorption (measured by CTX). GIP rapidly reduced CTX levels in the period from 45 to 120 min after injection, while GLP-2 had a more delayed effect with reduced CTX levels in the period from 120 to 240 min after injection. Combining GIP and GLP-2 showed complementary effects resulting in a sustained inhibition of CTX with reduced levels from 45 to 240 min after injection. Furthermore, GIP acutely increased bone formation (measured by P1NP). CONCLUSION: Both GIP and GLP-2 reduced CTX during the night and had complementary effects when combined.


Sujet(s)
Résorption osseuse , Glucagon-like peptide 2 , Glycémie , Remodelage osseux , Résorption osseuse/traitement médicamenteux , Femelle , Peptide gastrointestinal , Humains , Fragments peptidiques , Post-ménopause
13.
Eur J Case Rep Intern Med ; 8(3): 002352, 2021.
Article de Anglais | MEDLINE | ID: mdl-33869096

RÉSUMÉ

Sulfonylurea monotherapy is the standard treatment for patients with the most common form of permanent neonatal diabetes, KCNJ11 neonatal diabetes, but it is not always sufficient. For the first time, we present a case of successful use of a GLP-1 receptor agonist as add-on therapy in the treatment of a patient with KCNJ11 neonatal diabetes and insufficient effect of sulfonylurea monotherapy. Good glycaemic control was maintained with a HbA1c level of 48 mmol/mol (6.5%) at the end of 26 months' follow-up. LEARNING POINTS: Genetic testing is important in patients with neonatal diabetes.Sulfonylurea is the standard treatment for patients with the most common mutation (KCNJ11).We present the novel use of a GLP-1 receptor agonist as effective add-on therapy in a patient with KCNJ11 neonatal diabetes and insufficient effect of sulfonylurea monotherapy.

14.
Surg Obes Relat Dis ; 17(1): 55-63, 2021 Jan.
Article de Anglais | MEDLINE | ID: mdl-33039341

RÉSUMÉ

BACKGROUND: Postbariatric hypoglycemia (PBH) is a potentially serious complication after Roux-en-Y gastric bypass (RYGB), and impaired counterregulatory hormone responses have been suggested to contribute to the condition. OBJECTIVES: We evaluated counterregulatory responses during postprandial hypoglycemia in individuals with PBH who underwent RYGB. SETTING: University hospital. METHODS: Eleven women with documented PBH who had RYGB underwent a baseline liquid mixed meal test (MMT) followed by 5 MMTs preceded by treatment with (1) acarbose 50 mg, (2) sitagliptin 100 mg, (3) verapamil 120 mg, (4) liraglutide 1.2 mg, and (5) pasireotide 300 µg. Blood was collected at fixed time intervals. Plasma and serum were analyzed for glucose, insulin, glucagon, epinephrine, norepinephrine, pancreatic polypeptide (PP), and cortisol. RESULTS: During the baseline MMT, participants had nadir blood glucose concentrations of 3.3 ± .2 mmol/L. At the time of nadir glucose, there was a small but significant increase in plasma glucagon. Plasma epinephrine concentrations were not increased at nadir glucose but were significantly elevated by the end of the MMT. There were no changes in norepinephrine, PP, and cortisol concentrations in response to hypoglycemia. After treatment with sitagliptin, 8 individuals had glucose nadirs <3.2 mmol/L (versus 4 individuals at baseline), and significant increases in glucagon, PP, and cortisol responses were observed. CONCLUSIONS: In response to postprandial hypoglycemia, individuals with PBH who underwent RYGB only had minor increases in counterregulatory hormones, while larger hormone responses occurred when glucose levels were lowered during treatment with sitagliptin. The glycemic threshold for counterregulatory activation could be altered in individuals with PBH, possibly explained by recurrent hypoglycemia.


Sujet(s)
Dérivation gastrique , Hypoglycémie , Obésité morbide , Glycémie , Femelle , Dérivation gastrique/effets indésirables , Humains , Hypoglycémie/étiologie , Insuline , Obésité morbide/chirurgie
15.
Cell Rep Med ; 1(1): 100006, 2020 04 21.
Article de Anglais | MEDLINE | ID: mdl-33205056

RÉSUMÉ

Individuals with obesity due to pathogenic heterozygous melanocortin 4 receptor (MC4R) mutations can be treated efficiently with the glucagon-like peptide-1 receptor agonist (GLP-1 RA) liraglutide. Here, we report the effect of 16 weeks of liraglutide 3 mg/day treatment in a woman with morbid obesity and type 2 diabetes (T2D) due to homozygous pathogenic MC4R mutation. The body weight loss was 9.7 kg, similar to weight loss in heterozygous MC4R mutation carriers and common obesity. In addition, the treatment led to clinically relevant decreases in fasting glucose, triglycerides, systolic blood pressure, and normalization of glucose tolerance. We conclude that liraglutide reduces body weight and blood glucose levels in hetero- and homozygous MC4R mutation carriers. This serves as proof-of-concept that MC4Rs are not required for the body weight and glucose lowering effects of GLP-1 RAs and that liraglutide may be used as part of the treatment of obesity and T2D due to MC4R mutations.


Sujet(s)
Diabète de type 2/traitement médicamenteux , Liraglutide/usage thérapeutique , Obésité morbide/traitement médicamenteux , Récepteur de la mélanocortine de type 4/génétique , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Danemark , Diabète de type 2/complications , Diabète de type 2/génétique , Femelle , Récepteur du peptide-1 similaire au glucagon/agonistes , Homozygote , Humains , Adulte d'âge moyen , Mutation , Obésité morbide/complications , Obésité morbide/génétique , Perte de poids/effets des médicaments et des substances chimiques
16.
Am J Physiol Renal Physiol ; 318(6): F1409-F1417, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32390511

RÉSUMÉ

Glucagon-like peptide-1 (GLP-1) is an incretin hormone known to stimulate postprandial insulin release. However, GLP-1 also exerts extrapancreatic effects, including renal effects. Some of these renal effects are attenuated in hypertensive rats, where renal expression of GLP-1 receptors is reduced. Here, we assessed the expression and vascular function of GLP-1 receptors in kidneys from young prehypertensive rats. We also examined GLP-1-induced vasodilation in the renal vasculature in wild-type (WT) and GLP-1 receptor knockout mice using wire and pressure myography and the isolated perfused juxtamedullary nephron preparation. We investigated whether GLP-1 and the metabolite GLP-1(9-36)amide had renal vascular effects independent of the known GLP-1 receptor. We hypothesized that hypertension decreased expression of renal GLP-1 receptors. We also hypothesized that GLP-1-induced renal vasodilatation depended on expression of the known GLP-1 receptor. In contrast to normotensive rats, no immunohistochemical staining or vasodilatory function of GLP-1 receptors was found in kidneys from prehypertensive rats. In WT mice, GLP-1 induced renal vasodilation and reduced the renal autoregulatory response. The GLP-1 receptor antagonist exendin 9-39 inhibited relaxation, and GLP-1(9-36)amide had no vasodilatory effect. In GLP-1 receptor knockout mice, no relaxation induced by GLP-1 or GLP-1(9-36)amide was found, the autoregulatory response in afferent arterioles was normal, and no GLP-1-induced reduction of autoregulation was found. We conclude that in prehypertensive kidneys, expression and function of GLP-1 receptors is lost. The renal vasodilatory effect of GLP-1 is mediated exclusively by the known GLP-1 receptor. GLP-1(9-36)amide has no renal vasodilatory effect. GLP-1 attenuates renal autoregulation by reducing the myogenic response.


Sujet(s)
Artérioles/effets des médicaments et des substances chimiques , Pression sanguine/effets des médicaments et des substances chimiques , Glucagon-like peptide 1/pharmacologie , Récepteur du peptide-1 similaire au glucagon/agonistes , Rein/vascularisation , Préhypertension/métabolisme , Artère rénale/effets des médicaments et des substances chimiques , Vasodilatation/effets des médicaments et des substances chimiques , Vasodilatateurs/pharmacologie , Animaux , Artérioles/métabolisme , Artérioles/physiopathologie , Modèles animaux de maladie humaine , Femelle , Glucagon-like peptide 1/analogues et dérivés , Récepteur du peptide-1 similaire au glucagon/génétique , Récepteur du peptide-1 similaire au glucagon/métabolisme , Mâle , Souris de lignée C57BL , Souris knockout , Préhypertension/génétique , Préhypertension/physiopathologie , Rats de lignée SHR , Artère rénale/métabolisme , Artère rénale/physiopathologie
17.
Am J Physiol Endocrinol Metab ; 318(6): E956-E964, 2020 06 01.
Article de Anglais | MEDLINE | ID: mdl-32182123

RÉSUMÉ

Enhanced meal-related enteroendocrine secretion, particularly of glucagon-like peptide-1 (GLP-1), contributes to weight-loss and improved glycemia after Roux-en-Y gastric bypass (RYGB). Dietary glucose drives GLP-1 and glucose-dependent insulinotropic polypeptide (GIP) secretion postoperatively. Understanding how glucose triggers incretin secretion following RYGB could lead to new treatments of diabetes and obesity. In vitro, incretin release depends on glucose absorption via sodium-glucose cotransporter 1 (SGLT1). We investigated the importance of SGLT1/SGLT2 for enteropancreatic hormone concentrations and glucose metabolism after RYGB in a randomized, controlled, crossover study. Ten RYGB-operated patients ingested 50 g of oral glucose with and without acute pretreatment with 600 mg of the SGLT1/SGLT2-inhibitor canagliflozin. Paracetamol and 3-O-methyl-d-glucopyranose (3-OMG) were added to the glucose drink to evaluate rates of intestinal entry and absorption of glucose, respectively. Blood samples were collected for 4 h. The primary outcome was 4-h plasma GLP-1 (incremental area-under the curve, iAUC). Secondary outcomes included glucose, GIP, insulin, and glucagon. Canagliflozin delayed glucose absorption (time-to-peak 3-OMG: 50 vs. 132 min, P < 0.01) but did not reduce iAUC GLP-1 (6,067 vs. 7,273·min·pmol-1·L-1, P = 0.23), although peak GLP-1 concentrations were lowered (-28%, P = 0.03). Canagliflozin reduced GIP (iAUC -28%, P = 0.01; peak concentrations -57%, P < 0.01), insulin, and glucose excursions, whereas plasma glucagon (AUC 3,216 vs. 4,160 min·pmol·L-1, P = 0.02) and amino acids were increased. In conclusion, acute SGLT1/SGLT2-inhibition during glucose ingestion did not reduce 4-h plasma GLP-1 responses in RYGB-patients but attenuated the early rise in GLP-1, GIP, and insulin, whereas late glucagon concentrations were increased. The results suggest that SGLT1-mediated glucose absorption contributes to incretin hormone secretion after RYGB.


Sujet(s)
Canagliflozine/pharmacologie , Dérivation gastrique , Peptide gastrointestinal/métabolisme , Glucagon-like peptide 1/métabolisme , Transporteur-1 sodium-glucose/métabolisme , Inhibiteurs du cotransporteur sodium-glucose de type 2/pharmacologie , Transporteur-2 sodium-glucose/métabolisme , Glycémie/effets des médicaments et des substances chimiques , Glycémie/métabolisme , Peptide C/effets des médicaments et des substances chimiques , Peptide C/métabolisme , Études croisées , Peptide gastrointestinal/effets des médicaments et des substances chimiques , Glucagon/effets des médicaments et des substances chimiques , Glucagon/métabolisme , Glucagon-like peptide 1/effets des médicaments et des substances chimiques , Hyperglycémie provoquée , Humains , Incrétines/métabolisme , Insuline/métabolisme , Adulte d'âge moyen , Polypeptide pancréatique/effets des médicaments et des substances chimiques , Polypeptide pancréatique/métabolisme , Transporteur-1 sodium-glucose/antagonistes et inhibiteurs
18.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Article de Anglais | MEDLINE | ID: mdl-32077470

RÉSUMÉ

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


Sujet(s)
Glycémie/effets des médicaments et des substances chimiques , Métabolisme énergétique/effets des médicaments et des substances chimiques , Motilité gastrointestinale/effets des médicaments et des substances chimiques , Récepteur du peptide-1 similaire au glucagon/antagonistes et inhibiteurs , Récepteur hormone gastrointestinale/antagonistes et inhibiteurs , Adulte , Sujet âgé , Études croisées , Méthode en double aveugle , Vésicule biliaire/métabolisme , Peptide gastrointestinal/administration et posologie , Volontaires sains , Humains , Incrétines/sang , Mâle , Repas , Adulte d'âge moyen , Fragments peptidiques/administration et posologie , Période post-prandiale/effets des médicaments et des substances chimiques , Jeune adulte
19.
Physiol Rep ; 8(2): e14352, 2020 01.
Article de Anglais | MEDLINE | ID: mdl-31984675

RÉSUMÉ

Gastrin and cholecystokinin (CCK) are hormones released from endocrine cells in the antral stomach (gastrin), the duodenum, and the jejunum (CCK). Recent reports, based on secretion experiments in an enteroendocrine cell line (NCI-H716) and gastrin receptor expression in proglucagon-expressing cells from the rat colon, suggested that gastrin could be a regulator of glucagon-like peptide-1 (GLP-1) secretion. To investigate these findings, we studied the acute effects of CCK-8 (a CCK1/CCK2 (gastrin) receptor agonist) and gastrin-17 (a CCK2(gastrin) receptor agonist) in robust ex vivo models: the isolated perfused rat small intestine and the isolated perfused rat colon. Small intestines from Wistar rats (n = 6), were perfused intraarterially over 80 min. During the perfusion, CCK (1 nmol/L) and gastrin (1 nmol/L) were infused over 10-min periods separated by washout/baseline periods. Colons from Wistar rats (n = 6) were perfused intraarterially over 100 min. During the perfusion, CCK (1 nmol/L), vasoactive intestinal peptide (VIP) (10 nmol/L), and glucose-dependent insulinotropic polypeptide (GIP) (1 nmol/L) were infused over 10-min periods separated by washout/baseline periods. In the perfused rat small intestines neither CCK nor gastrin stimulated the release of GLP-1 or neurotensin. In the perfused rat colon, neither CCK or VIP stimulated GLP-1 or peptide YY (PYY) release, but GIP stimulated both GLP-1 and PYY release. In both sets of experiments, bombesin, a gastrin-releasing peptide analog, served as a positive control. Our findings do not support the suggestion that gastrin or CCK participate in the acute regulation of intestinal GLP-1 secretion, but that GIP may play a role in the regulation of hormone secretion from the colon.


Sujet(s)
Cholécystokinine/pharmacologie , Côlon/métabolisme , Gastrines/pharmacologie , Glucagon-like peptide 1/métabolisme , Intestin grêle/métabolisme , Neurotensine/métabolisme , Peptide YY/métabolisme , Animaux , Côlon/effets des médicaments et des substances chimiques , Intestin grêle/effets des médicaments et des substances chimiques , Mâle , Rats , Rat Wistar , Récepteur de la cholécystokinine de type B/agonistes , Récepteur cholécystokinine/agonistes , Peptide vasoactif intestinal/pharmacologie
20.
Diabetes Obes Metab ; 22(2): 141-148, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31468642

RÉSUMÉ

Type 2 diabetes mellitus (T2DM) is an increasingly prevalent chronic condition, characterized by abnormally elevated blood glucose concentrations and, as a consequence, increased risk of micro- and macrovascular complications. Metformin is usually the first-line glucose-lowering medication in T2DM; however, despite being used for more than 60 years, the mechanism underlying the glucose-lowering action of metformin remains incompletely understood. Although metformin reduces hepatic glucose production, there is persuasive evidence that the gastrointestinal tract is crucial in mediating this effect, particularly via secretion of the incretin hormone glucagon-like peptide 1 (GLP-1). It is now well recognized that bile acids, in addition to their established function in fat digestion and absorption, are important regulators of glucose metabolism. Exposure of the small and large intestine to bile acids induces GLP-1 secretion, modulates the composition of the gut microbiota, and reduces postprandial blood glucose excursions in humans with and without T2DM. Metformin reduces intestinal bile acid resorption substantially, such that intraluminal bile acids may, at least in part, account for its glucose-lowering effect. The present review focuses on the conceptual shift in our understanding as to how metformin lowers blood glucose in T2DM, with a particular emphasis on the role of intestinal bile acids.


Sujet(s)
Acides et sels biliaires/physiologie , Glycémie/effets des médicaments et des substances chimiques , Diabète de type 2/sang , Diabète de type 2/traitement médicamenteux , Metformine/pharmacologie , Acides et sels biliaires/pharmacologie , Glycémie/métabolisme , Métabolisme glucidique/effets des médicaments et des substances chimiques , Glucose/métabolisme , Humains , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/métabolisme , Intestins/effets des médicaments et des substances chimiques , Voies et réseaux métaboliques/effets des médicaments et des substances chimiques , Metformine/usage thérapeutique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE