Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.571
1.
Nutrients ; 16(9)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38732518

Vitamin D3 (VD3) is a steroid hormone that plays pivotal roles in pathophysiology, and 1,25(OH)2D3 is the most active form of VD3. In the current study, the crucial role of VD3 in maintaining energy homeostasis under short-term fasting conditions was investigated. Our results confirmed that glucose-depriving pathways were inhibited while glucose-producing pathways were strengthened in zebrafish after fasting for 24 or 48 h. Moreover, VD3 anabolism in zebrafish was significantly suppressed in a time-dependent manner under short-fasting conditions. After fasting for 24 or 48 h, zebrafish fed with VD3 displayed a higher gluconeogenesis level and lower glycolysis level in the liver, and the serum glucose was maintained at higher levels, compared to those fed without VD3. Additionally, VD3 augmented the expression of fatty acids (FAs) transporter cd36 and lipogenesis in the liver, while enhancing lipolysis in the dorsal muscle. Similar results were obtained in cyp2r1-/- zebrafish, in which VD3 metabolism is obstructed. Importantly, it was observed that VD3 induced the production of gut GLP-1, which is considered to possess a potent gluconeogenic function in zebrafish. Meanwhile, the gene expression of proprotein convertase subtilisin/kexin type 1 (pcsk1), a GLP-1 processing enzyme, was also induced in the intestine of short-term fasted zebrafish. Notably, gut microbiota and its metabolite acetate were involved in VD3-regulated pcsk1 expression and GLP-1 production under short-term fasting conditions. In summary, our study demonstrated that VD3 regulated GLP-1 production in zebrafish by influencing gut microbiota and its metabolite, contributing to energy homeostasis and ameliorating hypoglycemia under short-term fasting conditions.


Cholecalciferol , Energy Metabolism , Fasting , Homeostasis , Zebrafish , Animals , Cholecalciferol/metabolism , Cholecalciferol/pharmacology , Liver/metabolism , Gluconeogenesis , Gastrointestinal Microbiome/physiology , Blood Glucose/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/blood
2.
Clin Transl Sci ; 17(5): e13809, 2024 May.
Article En | MEDLINE | ID: mdl-38700326

DPP4 inhibitors are widely prescribed as treatments for type 2 diabetes. Because drug responses vary among individuals, we initiated investigations to identify genetic variants associated with the magnitude of drug responses. Sitagliptin (100 mg) was administered to 47 healthy volunteers. Several endpoints were measured to assess clinically relevant responses - including the effect of sitagliptin on glucose and insulin levels during an oral glucose tolerance test (OGTT). This pilot study confirmed that sitagliptin (100 mg) decreased the area under the curve for glucose during an OGTT (p = 0.0003). Furthermore, sitagliptin promoted insulin secretion during the early portion of the OGTT as reflected by an increase in the ratio of plasma insulin at 30 min divided by plasma insulin at 60 min (T30:T60) from mean ± SEM 0.87 ± 0.05 to 1.62 ± 0.36 mU/L (p = 0.04). The magnitude of sitagliptin's effect on insulin secretion (as judged by the increase in the T30:T60 ratio for insulin) was correlated with the magnitude of sitagliptin-induced increase in the area under the curve for intact plasma GLP1 levels during the first hour of the OGTT. This study confirmed previously reported sex differences in glucose and insulin levels during an OGTT. Specifically, females exhibited higher levels of glucose and insulin at the 90-180 min time points. However, we did not detect significant sex-associated differences in the magnitude of sitagliptin-induced changes in T30:T60 ratios for either glucose or insulin. In conclusion, T30:T60 ratios for insulin and glucose during an OGTT provide useful indices to assess pharmacodynamic responses to DPP4 inhibitors.


Blood Glucose , Glucose Tolerance Test , Insulin Secretion , Insulin , Sitagliptin Phosphate , Humans , Sitagliptin Phosphate/pharmacology , Sitagliptin Phosphate/administration & dosage , Male , Female , Adult , Insulin/blood , Insulin/metabolism , Insulin Secretion/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Blood Glucose/analysis , Young Adult , Dipeptidyl-Peptidase IV Inhibitors/administration & dosage , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Pilot Projects , Healthy Volunteers , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/blood , Middle Aged , Sex Factors
3.
Nutrients ; 16(8)2024 Apr 20.
Article En | MEDLINE | ID: mdl-38674919

This study investigates the effect of daily consumption of wheat biscuits enriched with plant proteins in postprandial metabolic responses of women with overweight/obesity who follow an energy-restricted diet. Thirty apparently healthy women participated in a 12-week randomized controlled trial and were assigned either to a control (CB) or an intervention (PB) group. Participants consumed daily either a conventional (CB) or an isocaloric wheat biscuit enriched with plant proteins (PB) containing high amounts of amino acids with appetite-regulating properties, i.e., BCAAs and L-arg. At baseline and the end of the intervention, a mixed meal tolerance test was performed. The responses of glucose, insulin, ghrelin, GLP-1, and glicentin were evaluated over 180 min. After 12 weeks, both groups experienced significant decreases in body weight, fat mass, and waist circumference. In the PB group, a trend towards higher weight loss was observed, accompanied by lower carbohydrate, fat, and energy intakes (p < 0.05 compared to baseline and CB group), while decreases in fasting insulin and the HOMA-IR index were also observed (p < 0.05 compared to baseline). In both groups, similar postprandial glucose, ghrelin, and GLP-1 responses were detected, while iAUC for insulin was lower (p < 0.05). Interestingly, the iAUC of glicentin was greater in the PB group (p < 0.05 compared to baseline). Subjective appetite ratings were beneficially affected in both groups (p < 0.05). Consumption of wheat biscuits enriched in plant proteins contributed to greater weight loss, lower energy intake, and insulin resistance and had a positive impact on postprandial glicentin response, a peptide that can potentially predict long-term weight loss and decreased food intake.


Blood Glucose , Obesity , Overweight , Postprandial Period , Triticum , Humans , Female , Adult , Obesity/diet therapy , Obesity/metabolism , Overweight/diet therapy , Overweight/metabolism , Blood Glucose/metabolism , Middle Aged , Insulin/blood , Plant Proteins/administration & dosage , Ghrelin/blood , Caloric Restriction/methods , Weight Loss , Energy Intake , Glucagon-Like Peptide 1/blood
4.
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
5.
Neurogastroenterol Motil ; 36(5): e14763, 2024 May.
Article En | MEDLINE | ID: mdl-38342974

BACKGROUND: Altered prandial glycemic response after Roux-en-Y gastric bypass (RYGB) is exaggerated in patients with post-RYGB hypoglycemia. Increased contribution of glucagon-like peptide 1 (GLP-1) to prandial insulin secretion plays a key role in developing hypoglycemia after RYGB, but the role of nonhormonal gut factors remains unknown. Here, the effect of vagal activation on prandial bile acid (BA) composition in relation to glucose, insulin and gut hormone responses was examined in a small size group of nondiabetic subjects after RYGB with intact gallbladder compared to nonoperated controls. METHODS: Concentrations of blood glucose, hormones, and BAs were measured in two RYGB subjects with documented hypoglycemia (HGB), three asymptomatic RYGB-treated subjects (AGB), and four nonoperated controls with intact gallbladders during a meal-tolerance test with (MTT-Sham) and without (MTT) preceding modified sham feeding (chew and spit). KEY RESULTS: Meal ingestion raised serum total BAs in RYGB-treated subjects without any effect in nonoperated controls. Modified sham feeding similarly increased meal-induced responses of conjugated BAs (CBAs) in all subjects (p < 0.05 compared to MTT alone), whereas unconjugated BAs (UBAs), mainly deoxycholic and chenodeoxycholic acid, were raised only in the HGB group (p < 0.001 for interaction). Prandial UBAs had an inverse correlation with glucose nadir (r = -0.75, p < 0.05) and were directly associated with ISR and GLP-1 during MTT-Sham. CONCLUSIONS & INFERENCES: In this small cohort, vagal activation by modified sham feeding increases prandial CBAs in both operated and nonoperated subjects but enhances UBAs only in patients with documented post-RYGB hypoglycemia. Our findings highlight a potential role for nonhormonal gut factors, such as BA and gut microbiome, in glucose abnormalities after RYGB.


Bile Acids and Salts , Blood Glucose , Gastric Bypass , Hypoglycemia , Vagus Nerve , Humans , Gastric Bypass/adverse effects , Bile Acids and Salts/blood , Blood Glucose/metabolism , Male , Female , Adult , Hypoglycemia/etiology , Hypoglycemia/blood , Middle Aged , Glucagon-Like Peptide 1/blood , Insulin/blood
6.
Br J Nutr ; 131(10): 1730-1739, 2024 May 28.
Article En | MEDLINE | ID: mdl-38287700

The aim of this study was to assess whether adding Ca2+ to aggregate or native forms of ß-lactoglobulin alters gut hormone secretion, gastric emptying rates and energy intake in healthy men and women. Fifteen healthy adults (mean ± sd: 9M/6F, age: 24 ± 5 years) completed four trials in a randomised, double-blind, crossover design. Participants consumed test drinks consisting of 30 g of ß-lactoglobulin in a native form with (NATIVE + MINERALS) and without (NATIVE) a Ca2+-rich mineral supplement and in an aggregated form both with (AGGREG + MINERALS) and without the mineral supplement (AGGREG). Arterialised blood was sampled for 120 min postprandially to determine gut hormone concentrations. Gastric emptying was determined using 13C-acetate and 13C-octanoate, and energy intake was assessed with an ad libitum meal at 120 min. A protein × mineral interaction effect was observed for total glucagon-like peptide-1 (GLP-1TOTAL) incremental AUC (iAUC; P < 0·01), whereby MINERALS + AGGREG increased GLP-1TOTAL iAUC to a greater extent than AGGREG (1882 ± 603 v. 1550 ± 456 pmol·l-1·120 min, P < 0·01), but MINERALS + NATIVE did not meaningfully alter the GLP-1 iAUC compared with NATIVE (1669 ± 547 v. 1844 ± 550 pmol·l-1·120 min, P = 0·09). A protein × minerals interaction effect was also observed for gastric emptying half-life (P < 0·01) whereby MINERALS + NATIVE increased gastric emptying half-life compared with NATIVE (83 ± 14 v. 71 ± 8 min, P < 0·01), whereas no meaningful differences were observed between MINERALS + AGGREG v. AGGREG (P = 0·70). These did not result in any meaningful changes in energy intake (protein × minerals interaction, P = 0·06). These data suggest that the potential for Ca2+ to stimulate GLP-1 secretion at moderate protein doses may depend on protein form. This study was registered at clinicaltrials.gov (NCT04659902).


Calcium, Dietary , Cross-Over Studies , Energy Intake , Gastric Emptying , Glucagon-Like Peptide 1 , Lactoglobulins , Humans , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Male , Female , Adult , Double-Blind Method , Young Adult , Lactoglobulins/metabolism , Calcium, Dietary/administration & dosage , Dietary Supplements , Postprandial Period , Calcium/metabolism
7.
Nutrients ; 14(19)2022 Sep 30.
Article En | MEDLINE | ID: mdl-36235718

Growth differentiation factor 15 (GDF15) is a stress signal that can be induced by protein restriction and is associated with reduced food intake. Anorexia of aging, insufficient protein intake as well as high GDF15 concentrations often occur in older age, but it is unknown whether GDF15 concentrations change acutely after meal ingestion and affect appetite in older individuals. After an overnight fast, appetite was assessed in older (n = 20; 73.7 ± 6.30 years) and younger (n = 20; 25.7 ± 4.39 years) women with visual analogue scales, and concentrations of circulating GDF15 and glucagon-like peptide-1 (GLP-1) were quantified before and at 1, 2 and 4 h after ingestion of either dextrose (182 kcal) or a mixed protein-rich meal (450 kcal). In response to dextrose ingestion, appetite increased in both older and younger women, whereas GDF15 concentrations increased only in the older group. In older women, appetite response was negatively correlated with the GDF15 response (rho = -0.802, p = 0.005). Following high-protein ingestion, appetite increased in younger women, but remained low in the old, while GDF15 concentrations did not change significantly in either age group. GLP-1 concentrations did not differ between age groups or test meals. In summary, acute GDF15 response differed between older and younger women. Associations of postprandial appetite and GDF15 following dextrose ingestion in older women suggest a reduced appetite response when the GDF15 response is high, thus supporting the proposed anorectic effects of high GDF15 concentrations.


Appetite , Dietary Proteins , Glucose , Growth Differentiation Factor 15 , Adult , Aged , Cross-Over Studies , Dietary Proteins/administration & dosage , Eating , Energy Intake , Female , Glucagon-Like Peptide 1/blood , Glucose/administration & dosage , Growth Differentiation Factor 15/blood , Humans , Postprandial Period , Young Adult
8.
Diabetes Obes Metab ; 24(10): 2017-2026, 2022 10.
Article En | MEDLINE | ID: mdl-35676803

AIMS: Sacubitril/valsartan is a neprilysin-inhibitor/angiotensin II receptor blocker used for the treatment of heart failure. Recently, a post-hoc analysis of a 3-year randomized controlled trial showed improved glycaemic control with sacubitril/valsartan in patients with heart failure and type 2 diabetes. We previously reported that sacubitril/valsartan combined with a dipeptidyl peptidase-4 inhibitor increases active glucagon-like peptide-1 (GLP-1) in healthy individuals. We now hypothesized that administration of sacubitril/valsartan with or without a dipeptidyl peptidase-4 inhibitor would lower postprandial glucose concentrations (primary outcome) in patients with type 2 diabetes via increased active GLP-1. METHODS: We performed a crossover trial in 12 patients with obesity and type 2 diabetes. A mixed meal was ingested following five respective interventions: (a) a single dose of sacubitril/valsartan; (b) sitagliptin; (c) sacubitril/valsartan + sitagliptin; (d) control (no treatment); and (e) valsartan alone. Glucose, gut and pancreatic hormone responses were measured. RESULTS: Postprandial plasma glucose increased by 57% (incremental area under the curve 0-240 min) (p = .0003) and increased peak plasma glucose by 1.7 mM (95% CI: 0.6-2.9) (p = .003) after sacubitril/valsartan compared with control, whereas postprandial glucose levels did not change significantly after sacubitril/valsartan + sitagliptin. Glucagon, GLP-1 and C-peptide concentrations increased after sacubitril/valsartan, but insulin and glucose-dependent insulinotropic polypeptide did not change. CONCLUSIONS: The glucose-lowering effects of long-term sacubitril/valsartan treatment reported in patients with heart failure and type 2 diabetes may not depend on changes in entero-pancreatic hormones. Neprilysin inhibition results in hyperglucagonaemia and this may explain the worsen glucose tolerance observed in this study. CLINICALTRIALS: gov (NCT03893526).


Aminobutyrates , Angiotensin Receptor Antagonists , Biphenyl Compounds , Blood Glucose , Diabetes Mellitus, Type 2 , Heart Failure , Hypoglycemic Agents , Neprilysin , Valsartan , Aged , Aminobutyrates/therapeutic use , Angiotensin Receptor Antagonists/therapeutic use , Biphenyl Compounds/therapeutic use , Blood Glucose/analysis , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Drug Combinations , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Heart Failure/complications , Heart Failure/drug therapy , Humans , Hypoglycemic Agents/therapeutic use , Male , Middle Aged , Neprilysin/antagonists & inhibitors , Sitagliptin Phosphate/therapeutic use , Tetrazoles/therapeutic use , Valsartan/therapeutic use
9.
Sci Rep ; 12(1): 2510, 2022 02 15.
Article En | MEDLINE | ID: mdl-35169165

It has not been elucidated whether incretins affect insulin clearance in type 2 diabetes (T2D). We aimed exploring possible associations between insulin clearance and endogenously secreted or exogenously administered incretins in T2D patients. Twenty T2D patients were studied (16 males/4 females, 59 ± 2 years (mean ± standard error), BMI = 31 ± 1 kg/m2, HbA1c = 7.0 ± 0.1%). Patients were treated with metformin, sitagliptin, metformin/sitagliptin combination, and placebo (randomized order). On each treatment period, oral and isoglycemic intravenous glucose infusion tests were performed (OGTT, IIGI, respectively). We also studied twelve T2D patients (9 males/3 females, 61 ± 3 years, BMI = 30 ± 1 kg/m2, HbA1c = 7.3 ± 0.4%) that underwent infusion of GLP-1(7-36)-amide, GIP, GLP-1/GIP combination, and placebo. Plasma glucose, insulin, C-peptide, and incretins were measured. Insulin clearance was assessed as insulin secretion to insulin concentration ratio. In the first study, we found OGTT/IIGI insulin clearance ratio weakly inversely related to OGTT/IIGI total GIP and intact GLP-1 (R2 = 0.13, p < 0.02). However, insulin clearance showed some differences between sitagliptin and metformin treatment (p < 0.02). In the second study we found no difference in insulin clearance following GLP-1 and/or GIP infusion (p > 0.5). Thus, our data suggest that in T2D there are no relevant incretin effects on insulin clearance. Conversely, different antidiabetic treatments may determine insulin clearance variations.


Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/administration & dosage , Glucagon-Like Peptide 1/administration & dosage , Hypoglycemic Agents/administration & dosage , Incretins/administration & dosage , Insulin Secretion/drug effects , Metformin/administration & dosage , Peptide Fragments/administration & dosage , Sitagliptin Phosphate/administration & dosage , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Drug Therapy, Combination/methods , Female , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test/methods , Humans , Hypoglycemic Agents/blood , Incretins/blood , Insulin/blood , Male , Middle Aged , Peptide Fragments/blood , Sitagliptin Phosphate/blood , Treatment Outcome
10.
Sci Rep ; 12(1): 1896, 2022 02 03.
Article En | MEDLINE | ID: mdl-35115614

Sodium-glucose cotransporter 2 inhibitor (SGLT2i) treatment is a therapeutic approach for type 2 diabetes mellitus (T2DM). Some reports have shown that SGLT2i treatment improves insulin resistance; however, few studies have evaluated insulin resistance by the glucose clamp method. Hepatic insulin clearance (HIC) is a new pathophysiological mechanism of T2DM. The effect of SGLT2i treatment on hepatic insulin clearance and insulin resistance is not well known. We investigated the effect of SGLT2i treatment on insulin resistance, insulin secretion, incretin levels, body composition, and hepatic insulin clearance. We conducted a meal tolerance test (MTT) and a hyperinsulinemic-euglycemic clamp test in 9 T2DM patients. Ipragliflozin (50 mg/day) was administered, and the MTT and clamp test were performed after 4 months. We calculated HIC as the postprandial C-peptide AUC-to-insulin AUC ratio. We also measured GLP-1, GIP, and glucagon levels during the MTT. Body weight and HbA1c were decreased, although not significantly, after 4 months of treatment. Postprandial glucose, fasting insulin and postprandial insulin were significantly decreased. Insulin resistance with the glucose clamp was not changed, but the HOMA-IR and insulin sensitivity indices were significantly improved. Incretin and glucagon levels were not changed. Hepatic insulin clearance was significantly increased, but whole-body insulin clearance was not changed. The FIB-4 index and fatty liver index were significantly reduced. The HOMA-beta and insulinogenic indices were not changed, but the C-peptide index was significantly increased. Although the number of patients was small, these results suggested that SGLT2i treatment improved liver function, decreased hepatic insulin resistance, and increased hepatic insulin clearance, despite the small weight reduction.


Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Glucosides/therapeutic use , Insulin Resistance , Insulin/blood , Liver/drug effects , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Thiophenes/therapeutic use , Biomarkers/blood , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Female , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Glucosides/adverse effects , Humans , Japan , Liver/metabolism , Male , Middle Aged , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Thiophenes/adverse effects , Time Factors , Treatment Outcome
11.
Clin Nutr ; 41(2): 517-525, 2022 02.
Article En | MEDLINE | ID: mdl-35016146

BACKGROUND: Lactate serves as an alternative energy fuel but is also an important signaling metabolite. We aimed to investigate whether oral lactate administration affects appetite-regulating hormones, slows gastric emptying rate, and dampens appetite. METHODS: Ten healthy male volunteers were investigated on two separate occasions: 1) following oral ingestion of D/L-Na-lactate and 2) following oral ingestion of isotonic iso-voluminous NaCl and intravenous iso-lactemic D/L-Na-lactate infusions. Appetite was evaluated by questionnaires and ad libitum meal tests were performed at the end of each study day. Gastric emptying rate was evaluated using the acetaminophen test. RESULTS: Plasma concentrations of growth differential factor 15 (GDF15, primary outcome) increased following oral and iv administration of lactate (p < 0.001) with no detectable difference between interventions (p = 0.15). Oral lactate administration lowered plasma concentrations of acylated ghrelin (p = 0.02) and elevated glucagon like peptide-1 (GLP-1, p = 0.045), insulin (p < 0.001), and glucagon (p < 0.001) compared with iv administration. Oral lactate administration slowed gastric emptying (p < 0.001), increased the feeling of being "full" (p = 0.008) and lowered the "anticipated future food intake" (p = 0.007) compared with iv administration. Food intake during the ad libitum meal test did not differ between the two study days. CONCLUSION: Oral lactate administration has a direct effect on the upper gastrointestinal tract, affecting gut hormone secretion, motility and appetite sensations which cannot be mediated through lactate in the systemic circulation alone. These data suggest that compounds rich in lactate may be useful in the treatment of metabolic disease. CLINICAL TRIAL REGISTRY NUMBER: NCT0429981, https://clinicaltrials.gov/ct2/show/NCT04299815.


Appetite Depressants/administration & dosage , Appetite/drug effects , Gastric Emptying/drug effects , Lactic Acid/administration & dosage , Administration, Intravenous , Administration, Oral , Adult , Eating/physiology , Gastrointestinal Hormones/blood , Ghrelin/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Growth Differentiation Factor 15/blood , Healthy Volunteers , Humans , Insulin/blood , Male , Young Adult
12.
J Clin Endocrinol Metab ; 107(1): 77-87, 2022 01 01.
Article En | MEDLINE | ID: mdl-34508600

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.


Colon/metabolism , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Lactulose/metabolism , Administration, Oral , Adult , Colon/microbiology , Cross-Over Studies , Fermentation , Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Healthy Volunteers , Humans , Intestinal Mucosa/microbiology , Lactulose/administration & dosage , Male , Peptide YY/blood , Peptide YY/metabolism , Young Adult
13.
Am J Clin Nutr ; 115(1): 284-297, 2022 01 11.
Article En | MEDLINE | ID: mdl-34555151

BACKGROUND: Evidence is emerging that interdaily meal pattern variability potentially affects response such as thermic effect of food (TEF), macronutrient metabolism, and appetite. OBJECTIVES: To investigate the effect of irregular meal pattern on TEF, glucose, insulin, lipid profile, and appetite regulation in women who are overweight or with obesity and confirmed insulin resistance. DESIGN: In a randomized crossover trial, 9 women [mean ± SD BMI (in kg/m2): 33.3 ± 3.1] with confirmed insulin resistance consumed a regular (14 d; 6 meals/d) and an irregular (14 d; 3-9 meals/d) meal pattern separated by a 14-d washout interval. Identical foods were provided during the interventions, and at the start and end of each meal pattern, participants attended the laboratory after an overnight fast. Energy expenditure, glucose, insulin, lipids, adiponectin, leptin, glucagon-like peptide 1 (GLP-1), peptide YY (PYY), and ghrelin were measured at baseline and for 3 h after consumption of a test drink, after which an ad libitum test meal was offered. Subjective appetite ratings were recorded before and after the test drink, after the ad libitum meal, and during the intervention. Continuous interstitial glucose monitoring was undertaken for 7 consecutive days during each intervention. RESULTS: TEF (over 3 h) was significantly lower postirregular intervention compared with postregular (97.7 ± 19.2 kJ*3 h in postregular visit and 76.7 ± 35.2 kJ*3 h in postirregular visit, paired t test, P = 0.048). Differences in HOMA-IR between the 2 interventions (3.3 ± 1.7 and 3.6 ± 1.6 in postregular and postirregular meal pattern, respectively) were not significant. Net incremental AUC for GLP-1 concentrations (over 3 h) for the postregular meal pattern were higher (864.9 ± 456.1 pmol/L*3 h) than the postirregular meal pattern (487.6 ± 271.7 pmol/L*3 h, paired t test, P = 0.005). CONCLUSIONS: Following a 14-d period of an irregular meal pattern, TEF was significantly less than following a regular meal pattern, potentially compromising weight management if sustained long term. This study was registered at www.clinicaltrials.gov as NCT02582606.


Appetite/physiology , Blood Glucose/metabolism , Meals/physiology , Overweight/physiopathology , Thermogenesis/physiology , Adiponectin/blood , Adolescent , Adult , Blood Glucose Self-Monitoring , Energy Metabolism , Feeding Behavior/physiology , Female , Ghrelin/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin , Insulin Resistance , Leptin/blood , Lipids/blood , Middle Aged , Obesity/blood , Obesity/physiopathology , Overweight/blood , Peptide YY/blood , Young Adult
14.
J Clin Endocrinol Metab ; 107(2): e756-e766, 2022 01 18.
Article En | MEDLINE | ID: mdl-34463768

CONTEXT: Whether Roux-en-Y gastric bypass (RYGB) and sleeve gastrectomy (SG) differentially affect postprandial gastrointestinal hormones and ß-cell function in type 2 diabetes remains unclear. OBJECTIVE: We aimed to compare gastrointestinal hormones and ß-cell function, assessed by an oral glucose tolerance test (OGTT) 5 weeks and 1 year after surgery, hypothesizing higher glucagon-like peptide-1 (GLP-1) levels and greater ß-cell response to glucose after RYGB than after SG. METHODS: This study was a randomized, triple-blind, single-center trial at a tertiary care center in Norway. The primary outcomes were diabetes remission and IVGTT-derived ß-cell function. Participants with obesity and type 2 diabetes were allocated (1:1) to RYGB or SG. We measured gastrointestinal hormone profiles and insulin secretion as ß-cell glucose sensitivity (ß-GS) derived from 180-minute OGTTs. RESULTS: Participants were 106 patients (67% women), mean (SD) age 48 (10) years. Diabetes remission rates at 1 year were higher after RYGB than after SG (77% vs 48%; P = 0.002). Incremental area under the curve (iAUC0-180) GLP-1 and ß-GS increased more after RYGB than after SG, with 1-year between-group difference 1173 pmol/L*min (95% CI, 569-1776; P = 0.0010) and 0.45 pmol/kg/min/mmol (95% CI, 0.15-0.75; P = 0.0032), respectively. After surgery, fasting and postprandial ghrelin levels were higher and decremental AUC0-180 ghrelin, iAUC0-180 glucose-dependent insulinotropic polypeptide, and iAUC0-60 glucagon were greater after RYGB than after SG. Diabetes remission at 1 year was associated with higher ß-GS and higher GLP-1 secretion. CONCLUSION: RYGB was associated with greater improvement in ß-cell function and higher postprandial GLP-1 levels than SG.


Diabetes Mellitus, Type 2/metabolism , Gastrectomy/statistics & numerical data , Gastric Bypass/statistics & numerical data , Glucagon-Like Peptide 1/blood , Insulin-Secreting Cells/metabolism , Obesity, Morbid/surgery , Adult , Blood Glucose/analysis , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Female , Follow-Up Studies , Gastrectomy/methods , Gastric Bypass/methods , Glucose Tolerance Test , Humans , Insulin/metabolism , Male , Middle Aged , Obesity, Morbid/blood , Obesity, Morbid/metabolism , Postprandial Period , Treatment Outcome
15.
J Clin Endocrinol Metab ; 107(1): e205-e213, 2022 01 01.
Article En | MEDLINE | ID: mdl-34410410

CONTEXT: Dietary fat and protein impact postprandial hyperglycemia in people with type 1 diabetes, but the underlying mechanisms are poorly understood. Glucoregulatory hormones are also known to modulate gastric emptying and may contribute to this effect. OBJECTIVE: Investigate the effects of fat and protein on glucagon-like peptide (GLP-1), glucagon-dependent insulinotropic polypeptide (GIP) and glucagon secretion. METHODS: 2 crossover euglycemic insulin clamp clinical trials at 2 Australian pediatric diabetes centers. Participants were 12-21 years (n = 21) with type 1 diabetes for ≥1 year. Participants consumed a low-protein (LP) or high-protein (HP) meal in Study 1, and low-protein/low-fat (LPLF) or high-protein/high-fat (HPHF) meal in Study 2, all containing 30 g of carbohydrate. An insulin clamp was used to maintain postprandial euglycemia and plasma glucoregulatory hormones were measured every 30 minutes for 5 hours. Data from both cohorts (n = 11, 10) were analyzed separately. The main outcome measure was area under the curve of GLP-1, GIP, and glucagon. RESULTS: Meals low in fat and protein had minimal effect on GLP-1, while there was sustained elevation after HP (80.3 ± 16.8 pmol/L) vs LP (56.9 ± 18.6), P = .016, and HPHF (103.0 ± 26.9) vs LPLF (69.5 ± 31.9) meals, P = .002. The prompt rise in GIP after all meals was greater after HP (190.2 ± 35.7 pmol/L) vs LP (152.3 ± 23.3), P = .003, and HPHF (258.6 ± 31.0) vs LPLF (151.7 ± 29.4), P < .001. A rise in glucagon was also seen in response to protein, and HP (292.5 ± 88.1 pg/mL) vs LP (182.8 ± 48.5), P = .010. CONCLUSION: The impact of fat and protein on postprandial glucose excursions may be mediated by the differential secretion of glucoregulatory hormones. Further studies to better understand these mechanisms may lead to improved personalized postprandial glucose management.


Biomarkers/blood , Blood Glucose/analysis , Diabetes Mellitus, Type 1/physiopathology , Dietary Fats/administration & dosage , Dietary Proteins/administration & dosage , Hyperglycemia/epidemiology , Meals , Adult , Australia/epidemiology , C-Peptide/blood , Cross-Over Studies , Female , Follow-Up Studies , Gastric Emptying , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Humans , Hyperglycemia/blood , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Insulin/blood , Male , Prognosis
16.
Neurogastroenterol Motil ; 34(1): e14210, 2022 01.
Article En | MEDLINE | ID: mdl-34378827

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.


Gastrectomy , Gastric Bypass , Neurotensin/blood , Obesity/surgery , Vagotomy, Truncal , Blood Glucose , Glucagon-Like Peptide 1/blood , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Liraglutide/administration & dosage , Liraglutide/therapeutic use , Obesity/blood , Obesity/drug therapy , Postprandial Period
17.
Expert Rev Anti Infect Ther ; 20(3): 373-381, 2022 Mar.
Article En | MEDLINE | ID: mdl-34348067

INTRODUCTION: Understanding the pathogenesis and risk factors to control the coronavirus disease 2019 (COVID-19) is necessary. Due to the importance of the inflammatory pathways in the pathogenesis of COVID-19 patients, evaluating the effects of anti-inflammatory medications is important. Glucagon-like peptide 1 receptor agonist (GLP-1 RA) is awell-known glucose-lowering agent with anti-inflammatory effects. AREAS COVERED: Resources were extracted from the PubMed database, using keywords such as glucagon-like peptide-1, GLP-1 RA, SARS-CoV-2, COVID-19, inflammation, in April2021. In this review, the effects of GLP-1RA in reducing inflammation and modifying risk factors of COVID-19 severe complications are discussed. However, GLP-1 is degraded by DPP-4 with aplasma half-life of about 2-5 minutes, which makes it difficult to measure GLP-1 plasma level in clinical settings. EXPERT OPINION: Since no definitive treatment is available for COVID-19 so far, determining promising targets to design and/or repurpose effective medications is necessary.


COVID-19 Drug Treatment , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor/agonists , Anti-Inflammatory Agents/therapeutic use , Glucagon-Like Peptide 1/blood , Humans , SARS-CoV-2
18.
Am J Physiol Endocrinol Metab ; 322(2): E165-E172, 2022 02 01.
Article En | MEDLINE | ID: mdl-34843659

Long-term sleep deprivation (SD) is a bad lifestyle habit, especially among specific occupational practitioners, characterized by circadian rhythm misalignment and abnormal sleep/wake cycles. SD is closely associated with an increased risk of metabolic disturbance, particularly obesity and insulin resistance. The incretin hormone, glucagon-like peptide-1 (GLP-1), is a critical insulin release determinant secreted by the intestinal L-cell upon food intake. Besides, the gut microbiota participates in metabolic homeostasis and regulates GLP-1 release in a circadian rhythm manner. As a commonly recognized intestinal probiotic, Bifidobacterium has various clinical indications regarding its curative effect. However, few studies have investigated the effect of Bifidobacterium supplementation on sleep disorders. In the present study, we explored the impact of long-term SD on the endocrine metabolism of rhesus monkeys and determined the effect of Bifidobacterium supplementation on the SD-induced metabolic status. Lipid concentrations, body weight, fast blood glucose, and insulin levels increased after SD. Furthermore, after 2 mo of long-term SD, the intravenous glucose tolerance test showed that the glucose metabolism was impaired and the insulin sensitivity decreased. Moreover, 1 mo of Bifidobacterium oral administration significantly reduced blood glucose and attenuated insulin resistance in rhesus macaques. Overall, our results suggested that Bifidobacterium might be used to alleviate SD-induced aberrant glucose metabolism and improve insulin resistance. Also, it might help in better understanding the mechanisms governing the beneficial effects of Bifidobacterium.NEW & NOTEWORTHY Our findings demonstrated that long-term sleep deprivation is closely associated with metabolic syndromes. Bifidobacterium administration showed a superior effect on insulin resistance caused by sleep deprivation. Overall, we provide prevention and treatment methods for long-term sleep deprivation, a bad lifestyle habit among specific occupational practitioners, such as irregular shift workers.


Bifidobacterium , Dietary Supplements , Insulin Resistance , Sleep Deprivation/complications , Sleep Deprivation/diet therapy , Animals , Blood Glucose/analysis , Blood Glucose/metabolism , Body Weight , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Circadian Rhythm , Disease Models, Animal , Gastric Inhibitory Polypeptide/blood , Glucagon-Like Peptide 1/blood , Glucose Tolerance Test , Incretins/blood , Insulin/blood , Macaca mulatta , Male , Sleep Deprivation/blood , Treatment Outcome , Triglycerides/blood
19.
Eur J Endocrinol ; 186(2): 207-221, 2022 Jan 06.
Article En | MEDLINE | ID: mdl-34863038

OBJECTIVE: Type 2 diabetes (T2D) pathophysiology includes fasting and postprandial hyperglucagonemia, which has been linked to hyperglycemia via increased endogenous glucose production (EGP). We used a glucagon receptor antagonist (LY2409021) and stable isotope tracer infusions to investigate the consequences of hyperglucagonemia in T2D. DESIGN: A double-blinded, randomized, placebo-controlled crossover study was conducted. METHODS: Ten patients with T2D and ten matched non-diabetic controls underwent two liquid mixed meal tests preceded by single-dose administration of LY2409021 (100 mg) or placebo. Double-tracer technique was used to quantify EGP. Antagonist selectivity toward related incretin receptors was determined in vitro. RESULTS: Compared to placebo, LY2409021 lowered the fasting plasma glucose (FPG) from 9.1 to 7.1 mmol/L in patients and from 5.6 to 5.0 mmol/L in controls (both P < 0.001) by mechanisms involving reduction of EGP. Postprandial plasma glucose excursions (baseline-subtracted area under the curve) were unaffected by LY2409021 in patients and increased in controls compared to placebo. Glucagon concentrations more than doubled during glucagon receptor antagonism. The antagonist interfered with both glucagon-like peptide 1 and glucose-dependent insulinotropic polypeptide receptors, complicating the interpretation of the postprandial data. CONCLUSIONS: LY2409021 lowered FPG concentrations but did not improve postprandial glucose tolerance after a meal in patients with T2D and controls. The metabolic consequences of postprandial hyperglucagonemia are difficult to evaluate using LY2409021 because of its antagonizing effects on the incretin receptors.


Biphenyl Compounds , Blood Glucose , Diabetes Mellitus, Type 2 , Postprandial Period , Receptors, Glucagon , Adult , Aged , Female , Humans , Male , Middle Aged , Biphenyl Compounds/therapeutic use , Blood Glucose/analysis , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Double-Blind Method , Fasting , Gastric Inhibitory Polypeptide/blood , Glucagon/blood , Glucagon-Like Peptide 1/blood , Receptors, Glucagon/antagonists & inhibitors
20.
J Clin Endocrinol Metab ; 107(1): e188-e204, 2022 01 01.
Article En | MEDLINE | ID: mdl-34479362

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.


Biomarkers/blood , Gastric Emptying , Glucose/metabolism , Meals , Pancreatic Hormones/blood , Adult , C-Peptide/blood , Cholecystokinin/blood , Follow-Up Studies , Glucagon/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin/blood , Male , Prognosis , Prospective Studies , Young Adult
...