Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
1.
Endocrinology ; 162(12)2021 12 01.
Article in English | MEDLINE | ID: mdl-34477204

ABSTRACT

Recent antecedent hypoglycemia is a known source of defective glucose counter-regulation in diabetes; the mechanisms perpetuating the cycle of progressive α-cell failure and recurrent hypoglycemia remain unknown. Somatostatin has been shown to suppress the glucagon response to acute hypoglycemia in rodent models of type 1 diabetes. We hypothesized that somatostatin receptor 2 antagonism (SSTR2a) would restore glucagon counterregulation and delay the onset of insulin-induced hypoglycemia in recurrently hypoglycemic, nondiabetic male rats. Healthy, male, Sprague-Dawley rats (n = 39) received bolus injections of insulin (10 U/kg, 8 U/kg, 5 U/kg) on 3 consecutive days to induce hypoglycemia. On day 4, animals were then treated with SSTR2a (10 mg/kg; n = 17) or vehicle (n = 12) 1 hour prior to the induction of hypoglycemia using insulin (5 U/kg). Plasma glucagon level during hypoglycemia was ~30% lower on day 3 (150 ± 75 pg/mL; P < .01), and 68% lower on day 4 in the vehicle group (70 ± 52 pg/mL; P < .001) compared with day 1 (219 ± 99 pg/mL). On day 4, SSTR2a prolonged euglycemia by 25 ± 5 minutes (P < .05) and restored the plasma glucagon response to hypoglycemia. Hepatic glycogen content of SSTR2a-treated rats was 35% lower than vehicle controls after hypoglycemia induction on day 4 (vehicle: 20 ± 7.0 vs SSTR2a: 13 ± 4.4 µmol/g; P < .01). SSTR2a treatment reverses the cumulative glucagon deficit resulting from 3 days of antecedent hypoglycemia in healthy rats. This reversal is associated with decreased hepatic glycogen content and delayed time to hypoglycemic onset. We conclude that recurrent hypoglycemia produces glucagon counterregulatory deficiency in healthy male rats, which can be improved by SSTR2a.


Subject(s)
Glucagon/metabolism , Hypoglycemia/metabolism , Peptides, Cyclic/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Glucagon/drug effects , Glucose/metabolism , Hormone Antagonists/pharmacology , Hypoglycemia/pathology , Liver Glycogen/metabolism , Male , Rats , Rats, Sprague-Dawley , Receptors, Somatostatin/antagonists & inhibitors , Recurrence
2.
Am J Physiol Regul Integr Comp Physiol ; 321(3): R303-R316, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34259034

ABSTRACT

Neonatal antibiotics administered to human infants initiate gut microbiota dysbiosis that may have long-term effects on body weight and metabolism. We examined antibiotic-induced adaptations in pancreatic islets of the piglet, a well-accepted model of human infant microbiota and pancreas development. Neonatal piglets randomized to amoxicillin [30 mg/kg body wt/day; n = 7, antibiotic (ANTI)] or placebo [vehicle control; n = 7, control (CON)] from postnatal day (PND)0-13 were euthanized at PND7, 14, and 49. The metabolic phenotype along with functional, immunohistological, and transcriptional phenotypes of the pancreatic islets were studied. The gut microbiome was characterized by 16S rRNA gene sequencing, and microbial metabolites and microbiome-sensitive host molecules were measured. Compared with CON, ANTI PND7 piglets had elevated transcripts of genes involved in glucagon-like peptide 1 ((GLP-1) synthesis or signaling in islets (P < 0.05) coinciding with higher plasma GLP-1 (P = 0.11), along with increased tumor necrosis factor α (Tnf) (P < 0.05) and protegrin 1 (Npg1) (P < 0.05). Antibiotic-induced relative increases in Escherichia, Coprococcus, Ruminococcus, Dehalobacterium, and Oscillospira of the ileal microbiome at PND7 normalized after antibiotic withdrawal. In ANTI islets at PND14, the expression of key regulators pancreatic and duodenal homeobox 1 (Pdx1), insulin-like growth factor-2 (Igf2), and transcription factor 7-like 2 (Tcf7l2) was downregulated, preceding a 40% reduction of ß-cell area (P < 0.01) and islet insulin content at PND49 (P < 0.05). At PND49, a twofold elevated plasma insulin concentration (P = 0.07) was observed in ANTI compared with CON. We conclude that antibiotic treatment of neonatal piglets elicited gut microbial changes accompanied by phasic alterations in key regulatory genes in pancreatic islets at PND7 and 14. By PND49, reduced ß-cell area and islet insulin content were accompanied by elevated nonfasted insulin despite normoglycemia, indicative of islet stress.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Glucagon-Like Peptide 1/metabolism , Insulin-Secreting Cells/drug effects , Animals , Gastrointestinal Microbiome/physiology , Glucagon/drug effects , Glucagon/metabolism , Insulin/blood , Insulin-Secreting Cells/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Swine
3.
J Diabetes Investig ; 12(12): 2162-2171, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34022121

ABSTRACT

AIMS/INTRODUCTION: Differences in the glucose-lowering mechanisms of glucagon-like peptide-1 receptor agonists (GLP-1RAs) have been noted. Clarifying these differences could facilitate the choice of optimal drugs for individuals with type 2 diabetes and requires investigation in a clinical setting. MATERIALS AND METHODS: A single-arm, prospective, observational study was conducted to evaluate the effects of various GLP-1RAs on postprandial glucose excursion, secretions of insulin and glucagon as well as on the gastric emptying rate. Participants were subjected to meal tolerance tests before and 2 weeks and 12 weeks after GLP-1RA initiation. Effects on postprandial secretions of glucose-dependent insulinotropic polypeptide (GIP) and apolipoprotein B48 were also investigated. RESULTS: Eighteen subjects with type 2 diabetes received one of three GLP-1RAs, i.e., lixisenatide, n = 7; liraglutide, n = 6; or dulaglutide, n = 5. While 12-week administration of all of the GLP-1RAs significantly reduced HbA1c, only lixisenatide and liraglutide, but not dulaglutide, significantly reduced body weight. Postprandial glucose elevation was improved by all of the GLP-1RAs. Postprandial insulin levels were suppressed by lixisenatide, while insulin levels were enhanced by liraglutide. Postprandial glucagon levels were suppressed by lixisenatide. The gastric emptying rate was significantly delayed by lixisenatide, while liraglutide and dulaglutide had limited effects on gastric emptying. GIP secretion was suppressed by lixisenatide and liraglutide. Apolipoprotein B48 secretion was suppressed by all of the GLP-1RAs. CONCLUSIONS: All of the GLP-1RAs were found to improve HbA1c in a 12-week prospective observational study in Japanese individuals with type 2 diabetes. However, differences in the mechanisms of the glucose-lowering effects and body weight reduction were observed.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastric Emptying/drug effects , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacology , Adult , Apolipoprotein B-48/metabolism , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/blood , Female , Gastric Inhibitory Polypeptide/metabolism , Glucagon/drug effects , Glucagon-Like Peptides/analogs & derivatives , Glucagon-Like Peptides/pharmacology , Humans , Immunoglobulin Fc Fragments/pharmacology , Insulin/blood , Japan , Liraglutide/pharmacology , Male , Middle Aged , Peptides/pharmacology , Postprandial Period/drug effects , Prospective Studies , Recombinant Fusion Proteins/pharmacology
4.
Cardiovasc Drugs Ther ; 35(3): 491-503, 2021 06.
Article in English | MEDLINE | ID: mdl-32770520

ABSTRACT

PURPOSE: The glucose-lowering drug metformin has recently been shown to reduce myocardial oxygen consumption and increase myocardial efficiency in chronic heart failure (HF) patients without diabetes. However, it remains to be established whether these beneficial myocardial effects are associated with metformin-induced alterations in whole-body insulin sensitivity and substrate metabolism. METHODS: Eighteen HF patients with reduced ejection fraction and without diabetes (median age, 65 (interquartile range 55-68); ejection fraction 39 ± 6%; HbA1c 5.5 to 6.4%) were randomized to receive metformin (n = 10) or placebo (n = 8) for 3 months. We studied the effects of metformin on whole-body insulin sensitivity using a two-step hyperinsulinemic euglycemic clamp incorporating isotope-labeled tracers of glucose, palmitate, and urea. Substrate metabolism and skeletal muscle mitochondrial respiratory capacity were determined by indirect calorimetry and high-resolution respirometry, and body composition was assessed by bioelectrical impedance analysis. The primary outcome measure was change in insulin sensitivity. RESULTS: Compared with placebo, metformin treatment lowered mean glycated hemoglobin levels (absolute mean difference, - 0.2%; 95% CI - 0.3 to 0.0; p = 0.03), reduced body weight (- 2.8 kg; 95% CI - 5.0 to - 0.6; p = 0.02), and increased fasting glucagon levels (3.2 pmol L-1; 95% CI 0.4 to 6.0; p = 0.03). No changes were observed in whole-body insulin sensitivity, endogenous glucose production, and peripheral glucose disposal or oxidation with metformin. Equally, resting energy expenditure, lipid and urea turnover, and skeletal muscle mitochondrial respiratory capacity remained unaltered. CONCLUSION: Increased myocardial efficiency during metformin treatment is not mediated through improvements in insulin action in HF patients without diabetes. CLINICAL TRIAL REGISTRATION: URL: https://clinicaltrials.gov . Unique identifier: NCT02810132. Date of registration: June 22, 2016.


Subject(s)
Body Weight/drug effects , Heart Failure/drug therapy , Heart Failure/physiopathology , Insulin Resistance/physiology , Metformin/pharmacology , Aged , Body Composition , Calorimetry, Indirect , Double-Blind Method , Female , Glucagon/drug effects , Glycated Hemoglobin/drug effects , Humans , Male , Middle Aged , Mitochondria/drug effects , Muscle, Skeletal/drug effects , Oxygen Consumption/drug effects , Stroke Volume/drug effects
5.
Mol Metab ; 42: 101071, 2020 12.
Article in English | MEDLINE | ID: mdl-32896668

ABSTRACT

OBJECTIVE: Sodium-glucose cotransporter 2 (SGLT2) inhibitors (SGLT2i), or gliflozins, are anti-diabetic drugs that lower glycemia by promoting glucosuria, but they also stimulate endogenous glucose and ketone body production. The likely causes of these metabolic responses are increased blood glucagon levels, and decreased blood insulin levels, but the mechanisms involved are hotly debated. This study verified whether or not SGLT2i affect glucagon and insulin secretion by a direct action on islet cells in three species, using multiple approaches. METHODS: We tested the in vivo effects of two selective SGLT2i (dapagliflozin, empagliflozin) and a SGLT1/2i (sotagliflozin) on various biological parameters (glucosuria, glycemia, glucagonemia, insulinemia) in mice. mRNA expression of SGLT2 and other glucose transporters was assessed in rat, mouse, and human FACS-purified α- and ß-cells, and by analysis of two human islet cell transcriptomic datasets. Immunodetection of SGLT2 in pancreatic tissues was performed with a validated antibody. The effects of dapagliflozin, empagliflozin, and sotagliflozin on glucagon and insulin secretion were assessed using isolated rat, mouse and human islets and the in situ perfused mouse pancreas. Finally, we tested the long-term effect of SGLT2i on glucagon gene expression. RESULTS: SGLT2 inhibition in mice increased the plasma glucagon/insulin ratio in the fasted state, an effect correlated with a decline in glycemia. Gene expression analyses and immunodetections showed no SGLT2 mRNA or protein expression in rodent and human islet cells, but moderate SGLT1 mRNA expression in human α-cells. However, functional experiments on rat, mouse, and human (29 donors) islets and the in situ perfused mouse pancreas did not identify any direct effect of dapagliflozin, empagliflozin or sotagliflozin on glucagon and insulin secretion. SGLT2i did not affect glucagon gene expression in rat and human islets. CONCLUSIONS: The data indicate that the SGLT2i-induced increase of the plasma glucagon/insulin ratio in vivo does not result from a direct action of the gliflozins on islet cells.


Subject(s)
Glucagon/metabolism , Insulin Secretion/physiology , Sodium-Glucose Transporter 2/metabolism , Animals , Benzhydryl Compounds/pharmacology , Blood Glucose/metabolism , Glucagon/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Secreting Cells/metabolism , Glucose/metabolism , Glucosides/pharmacology , Humans , Insulin/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Mice , Pancreas/metabolism , Rats , Sodium-Glucose Transporter 2/physiology , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
Am J Physiol Endocrinol Metab ; 319(1): E133-E145, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32459527

ABSTRACT

Bromocriptine mesylate treatment was examined in dogs fed a high fat diet (HFD) for 8 wk. After 4 wk on HFD, daily bromocriptine (Bromo; n = 6) or vehicle (CTR; n = 5) injections were administered. Oral glucose tolerance tests were performed before beginning HFD (OGTT1), 4 wk after HFD began (Bromo only), and after 7.5 wk on HFD (OGTT3). After 8 wk on HFD, clamp studies were performed, with infusion of somatostatin and intraportal replacement of insulin (4× basal) and glucagon (basal). From 0 to 90 min (P1), glucose was infused via peripheral vein to double the hepatic glucose load; and from 90 to 180 min (P2), glucose was infused via the hepatic portal vein at 4 mg·kg-1·min-1, with the HGL maintained at 2× basal. Bromo decreased the OGTT glucose ΔAUC0-30 and ΔAUC0-120 by 62 and 27%, respectively, P < 0.05 for both) without significantly altering the insulin response. Bromo dogs exhibited enhanced net hepatic glucose uptake (NHGU) compared with CTR (~33 and 21% greater, P1 and P2, respectively, P < 0.05). Nonhepatic glucose uptake (non-HGU) was increased ~38% in Bromo in P2 (P < 0.05). Bromo vs. CTR had higher (P < 0.05) rates of glucose infusion (36 and 30%) and non-HGU (~40 and 27%) than CTR during P1 and P2, respectively. In Bromo vs. CTR, hepatic 18:0/16:0 and 16:1/16:0 ratios tended to be elevated in triglycerides and were higher (P < 0.05) in phospholipids, consistent with a beneficial effect of bromocriptine on liver fat accumulation. Thus, bromocriptine treatment improved glucose disposal in a glucose-intolerant model, enhancing both NHGU and non-HGU.


Subject(s)
Blood Glucose/drug effects , Bromocriptine/pharmacology , Diet, High-Fat , Dopamine Agonists/pharmacology , Glucose Intolerance/metabolism , Liver/drug effects , Animals , Blood Glucose/metabolism , Dogs , Fatty Acids, Nonesterified/metabolism , Glucagon/drug effects , Glucagon/metabolism , Glucose/metabolism , Glucose Clamp Technique , Glucose Tolerance Test , Glycogen/metabolism , Hepatic Veins , Insulin/metabolism , Lactic Acid/metabolism , Liver/metabolism , Portal Vein , Somatostatin
7.
Medicine (Baltimore) ; 99(14): e19685, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32243407

ABSTRACT

The effect of intensive insulin therapy on hyperglucagonemia in newly diagnosed type 2 diabetes (T2DM), and its associations with ß-cell function, has not been elucidated. This study assessed the effect of 12 weeks of intensive insulin therapy on hyperglucagonemia in newly diagnosed T2DM and its associations with ß-cell function, with reference to the effects of 12 weeks of oral hypoglycemic agents (OHAs).One hundred eight patients with newly diagnosed T2DM were enrolled from January 2015 to December 2015. The patients were randomly divided to receive, for 12 weeks, either intensive insulin therapy or OHAs. Meal tolerance tests were conducted at baseline before treatment (0 week), at 12 weeks (end of treatment), and 12 months after the initiation of treatment. The levels of glucagon, proinsulin, C-peptide (CP), and blood glucose were measured at timepoints 0, 30, and 120 minutes during the meal tolerance test.Intensive insulin treatment was associated with a decrease in glucagon levels (at 0, 30, and 120 minutes) and proinsulin/CP, and an increase in the insulin-secretion index ΔCP30/ΔG30 and ΔCP120/ΔG120, at 12 weeks and 12 months during the follow-up, compared with the corresponding effects of OHAs. Intensive insulin therapy could reduce but failed to normalize glucagon levels at 12 weeks. There were no correlations between the change of percentages in total area under the curve of glucagon and other glycemic parameters (proinsulin/CP; ΔCP30/ΔG30; or ΔCP120/ΔG120). Patients who received intensive insulin therapy were more likely to achieve their target glycemic goal and remission, compared with those who received OHAs.Short-term intensive insulin therapy facilitates the improvement of both ß-cell and α-cell function in newly diagnosed T2DM mellitus. Decline of ß-cell secretion and concomitant α-cell dysfunction may both be involved in the pathogenesis of T2DM.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Glucagon-Secreting Cells/drug effects , Hypoglycemic Agents/administration & dosage , Insulin/administration & dosage , Adult , Blood Glucose/drug effects , Carbohydrate Metabolism, Inborn Errors/blood , Carbohydrate Metabolism, Inborn Errors/etiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Drug Administration Schedule , Female , Glucagon/blood , Glucagon/drug effects , Glucose Tolerance Test , Glycated Hemoglobin/drug effects , Humans , Insulin-Secreting Cells/drug effects , Male , Middle Aged , Prospective Studies , Treatment Outcome
8.
Am J Physiol Endocrinol Metab ; 318(6): E956-E964, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32182123

ABSTRACT

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.


Subject(s)
Canagliflozin/pharmacology , Gastric Bypass , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Sodium-Glucose Transporter 1/metabolism , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Sodium-Glucose Transporter 2/metabolism , Blood Glucose/drug effects , Blood Glucose/metabolism , C-Peptide/drug effects , C-Peptide/metabolism , Cross-Over Studies , Gastric Inhibitory Polypeptide/drug effects , Glucagon/drug effects , Glucagon/metabolism , Glucagon-Like Peptide 1/drug effects , Glucose Tolerance Test , Humans , Incretins/metabolism , Insulin/metabolism , Middle Aged , Pancreatic Polypeptide/drug effects , Pancreatic Polypeptide/metabolism , Sodium-Glucose Transporter 1/antagonists & inhibitors
9.
Diabetes Res Clin Pract ; 158: 107892, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31669625

ABSTRACT

AIMS: Glucagon has an important role in glucose homeostasis. Recently, a new plasma glucagon assay based on liquid chromatography-high resolution mass spectrometry was developed. We evaluated the influence of a dipeptidyl peptidase-4 inhibitor (anagliptin) on plasma glucagon levels in Japanese patients with type 2 diabetes by using this new assay. METHODS: Twenty-four patients with type 2 diabetes were enrolled in a prospective, single-center, randomized, open-label study and were randomly allocated to 4 weeks of treatment with metformin (1000 mg/day) or anagliptin (200 mg/day). A liquid test meal labeled with sodium [13C] acetate was ingested before and after the treatment period. Samples of blood and expired air were collected over 3 h. Plasma levels of glucose, glucagon, C-peptide, glucagon-like peptide-1 (GLP-1), and glucose-dependent insulinotropic polypeptide (GIP) were measured, and gastric emptying was also evaluated. RESULTS: Twenty-two patients completed the study (metformin group: n = 10; anagliptin group: n = 12). Glycemic control showed similar improvement in both groups. In the anagliptin group, there was a slight decrease of the incremental area under the plasma concentration versus time curve for glucagon after the test meal (P = 0.048). In addition, the plasma level of active GLP-1 and GIP was increased, and plasma C-peptide was also increased versus baseline. Neither anagliptin nor metformin delayed gastric emptying. CONCLUSIONS: In patients with type 2 diabetes maintained endogenous insulin secretion, anagliptin increased the plasma level of active GLP-1 and GIP in association with a slight stimulation of insulin secretion and slight inhibition of glucagon secretion, but did not delay gastric emptying. Clinical Trial Registry: University hospital Medical Information Network UMIN000028293.


Subject(s)
Gastric Emptying/drug effects , Glucagon/drug effects , Hypoglycemic Agents/therapeutic use , Mass Spectrometry/methods , Metformin/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Diabetes Mellitus, Type 2/blood , Female , Humans , Hypoglycemic Agents/pharmacology , Male , Metformin/pharmacology , Middle Aged , Prospective Studies , Pyrimidines/pharmacology , Young Adult
10.
Cell Rep ; 28(6): 1447-1454.e4, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31390560

ABSTRACT

The newest classes of anti-diabetic agents include sodium-glucose cotransporter 2 (SGLT2) inhibitors and glucagon-like peptide 1 receptor (GLP1R) agonists. The SGLT2 inhibitor dapagliflozin reduces glucotoxicity by glycosuria but elevates glucagon secretion. The GLP1R agonist liraglutide inhibits glucagon; therefore, we hypothesize that the cotreatment of dapagliflozin with liraglutide could reduce hyperglucagonemia and hyperglycemia. Here we use five complementary models: human islet cultures, healthy mice, db/db mice, diet-induced obese (DIO) mice, and somatostatin receptor-2 (SSTR2) KO mice. A single administration of liraglutide and dapagliflozin in combination improves glycemia and reduces dapagliflozin-induced glucagon secretion in diabetic mice. Chronic treatment with liraglutide and dapagliflozin produces a sustainable reduction of glycemia compared with each drug alone. Moreover, liraglutide reduces dapagliflozin-induced glucagon secretion by enhancing somatostatin release, as demonstrated by SSTR2 inhibition in human islets and in mice. Collectively, these data provide mechanistic insights into how intra-islet GLP1R activation is critical for the regulation of glucose homeostasis.


Subject(s)
Benzhydryl Compounds/adverse effects , Diabetes Mellitus, Experimental/drug therapy , Glucagon/drug effects , Glucosides/adverse effects , Liraglutide/therapeutic use , Somatostatin/drug effects , Animals , Humans , Liraglutide/pharmacology , Male , Mice
11.
Brain Behav ; 9(9): e01340, 2019 09.
Article in English | MEDLINE | ID: mdl-31392839

ABSTRACT

INTRODUCTION: Ginseng polysaccharide (GPS, same as Panax polysaccharide) is a kind of polysaccharide extracted from ginseng. It has been reported that GPS has the ability to activate innate immunity, regulates blood sugar balance, and improves antioxidant capacity, but the effect on feeding behavior and its mechanism remains unclear. METHOD: To investigate the possible effect of GPS on feeding behavior of animals, mice were supplied with GPS in water, and food intake, hedonic feeding behavior, anxiety-like behavior, expression of appetite-regulation peptides in the central nervous system and glucose-related hormone levels in the serum of mice were measured. RESULTS: Ginseng polysaccharide significantly increased the average daily food intake in mice and promoted hedonic eating behavior. Meanwhile, the levels of serum glucose and glucagon were significantly reduced by GPS, and GPS promoted hypothalamic neuropeptide Y expression, inhibited proopiomelanocortin (POMC) expression, and reduced dopamine D1 receptor (DRD1) levels in the midbrain. We also found that the anxiety level of mice was significantly lower after GPS intake. In conclusion, oral supplementation with GPS promoted food intake in mice, most likely through the regulation of circulating glucose levels.


Subject(s)
Feeding Behavior/drug effects , Panax , Polysaccharides/pharmacology , Animals , Anxiety , Behavior, Animal/drug effects , Blood Glucose/drug effects , Blood Glucose/metabolism , Dietary Supplements , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine Plasma Membrane Transport Proteins/genetics , Eating/drug effects , Glucagon/drug effects , Glucagon/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Insulin/metabolism , Male , Mesencephalon/drug effects , Mesencephalon/metabolism , Mice , Neuropeptide Y/drug effects , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/drug effects , Pro-Opiomelanocortin/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/genetics , Receptors, Dopamine D2/drug effects , Receptors, Dopamine D2/genetics
12.
Eur J Endocrinol ; 181(2): 161-171, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31176298

ABSTRACT

OBJECTIVES: The aim of the study was to explore the role of GLP-1 receptor activation on the counter-regulation and symptoms of hypoglycemia in subjects who have undergone gastric bypass surgery (GBP). DESIGN: Experimental hyperinsulinemic-hypoglycemic clamp study. METHODS: Twelve post-GBP subjects participated in a randomized cross-over study with two hyperinsulinemic, hypoglycemic clamps (glucose nadir 2.7 mmol/L) performed on separate days with concomitant infusions of the GLP-1 analog exenatide or with saline, respectively. Continuous measurements of metabolites and counter-regulatory hormones as well as assessments of heart rate variability and symptoms of hypoglycemia were performed throughout the clamps. RESULTS: No effect of GLP-1 receptor activation on counter-regulatory hormones (glucagon, catecholamines, cortisol, GH) or glucose infusion rate was seen, but we found indications of a downregulation of the sympathetic relative to the parasympathetic nerve activity, as reflected in heart rate variability. No significant differences in symptom of hypoglycemia were observed. CONCLUSIONS/INTERPRETATION: Short-term exposure to a GLP-1 receptor agonist does not seem to impact the counter-regulatory hormonal and metabolic responses in post-GBP subjects during hypoglycemic conditions, suggesting that the improvement in symptomatic hypoglycemia post-GBP seen following treatment with GLP-1 receptor agonists may be mediated by mechanism not directly involved in counter-regulation.


Subject(s)
Blood Glucose/drug effects , Exenatide/pharmacology , Gastric Bypass , Heart Rate/drug effects , Hypoglycemia/metabolism , Incretins/pharmacology , Adult , Blood Glucose/metabolism , Catecholamines/metabolism , Cross-Over Studies , Female , Glucagon/drug effects , Glucagon/metabolism , Glucagon-Like Peptide 1/metabolism , Glucose Clamp Technique , Human Growth Hormone/drug effects , Human Growth Hormone/metabolism , Humans , Hydrocortisone/metabolism , Male , Middle Aged , Parasympathetic Nervous System/drug effects , Postoperative Period , Sympathetic Nervous System/drug effects
13.
Am J Physiol Endocrinol Metab ; 316(4): E660-E673, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30807215

ABSTRACT

Glucagon and insulin are important regulators of blood glucose. The importance of insulin receptor signaling for alpha-cell secretion and of glucagon receptor signaling for beta-cell secretion is widely discussed and of clinical interest. Amino acids are powerful secretagogues for both hormones, and glucagon controls amino acid metabolism through ureagenesis. The role of insulin in amino acid metabolism is less clear. Female C57BL/6JRj mice received an insulin receptor antagonist (IRA) (S961; 30 nmol/kg), a glucagon receptor antagonist (GRA) (25-2648; 100 mg/kg), or both GRA and IRA (GRA + IRA) 3 h before intravenous administration of similar volumes of saline, glucose (0.5 g/kg), or amino acids (1 µmol/g) while anesthetized with isoflurane. IRA caused basal hyperglycemia, hyperinsulinemia, and hyperglucagonemia. Unexpectedly, IRA lowered basal plasma concentrations of amino acids, whereas GRA increased amino acids, lowered glycemia, and increased glucagon but did not influence insulin concentrations. After administration of GRA + IRA, insulin secretion was significantly reduced compared with IRA administration alone. Blood glucose responses to a glucose and amino acid challenge were similar after vehicle and GRA + IRA administration but greater after IRA and lower after GRA. Anesthesia may have influenced the results, which otherwise strongly suggest that both hormones are essential for the maintenance of glucose homeostasis and that the secretion of both is regulated by powerful negative feedback mechanisms. In addition, insulin limits glucagon secretion, while endogenous glucagon stimulates insulin secretion, revealed during lack of insulin autocrine feedback. Finally, glucagon receptor signaling seems to be of greater importance for amino acid metabolism than insulin receptor signaling.


Subject(s)
Amino Acids/metabolism , Blood Glucose/metabolism , Glucagon/metabolism , Receptor, Insulin/metabolism , Receptors, Glucagon/metabolism , Amino Acids/drug effects , Animals , Blood Glucose/drug effects , Glucagon/drug effects , Glucose/metabolism , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Mice , Peptides/pharmacology , Receptor, Insulin/antagonists & inhibitors , Receptors, Glucagon/antagonists & inhibitors
14.
Zhonghua Nei Ke Za Zhi ; 58(1): 33-38, 2019 Jan 01.
Article in Chinese | MEDLINE | ID: mdl-30605948

ABSTRACT

Objective: To investigate the effect of liraglutide on glucagon release in obese type 2 diabetes (T2DM). Methods: A multi-center, prospective, and self-comparison study was conducted in four hospitals in Qingdao. Twenty-four patients with T2DM were selected and treated with liraglutide for 12 weeks. Glucagon levels before and after treatment were detected before and 30 min, 60 min and 120 min after meals. Results: After 12 weeks of treatment, the overall level of glucagon decreased, in which the differences in glucagon levels at 30 min [(220±79) ng/L vs. (203±77) ng/L, P<0.05] and 60 min [(248±119) ng/L vs. (203±82)ng/L, P<0.05] reached significance, respectively, comparing to those before treatment. The area under the curve of glucagon after treatment was significantly lower than that before treatment (438±190 vs. 389±153, P<0.05). In contrast, after treatment, the overall level of C-peptide increased, especially the levels at 30 min [(1.53±1.02) nmol/L vs.(2.03±1.29) nmol/L], 60 min [(1.93±1.19) nmol/L vs. (2.48±1.75) nmol/L] and 120 min [(2.36±1.47) nmol/L vs. (2.96±1.84) nmol/L], all P<0.05. The area under C-peptide curve increased significantly (3.6±2.2 vs. 4.6±2.9, P<0.05). Fasting plasma glucose, postprandial 2 h plasma glucose and glycosylated hemoglobin A1c were all lower than before, and the differences were statistically significant (P<0.05). Waist circumference and body mass index were significantly lower than before (P<0.05). The amount of insulin used for the treatment decreased by approximately 55.1% compared with that before liraglutide, and the difference was statistically significant (P<0.05). Conclusions: Liraglutide inhibits glucagon secretion and lowers blood glucose. It can also reduce body weight, improve islet cell function and reduce insulin use in T2DM.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/drug therapy , Glucagon/drug effects , Glycated Hemoglobin/metabolism , Hypoglycemic Agents/administration & dosage , Liraglutide/pharmacology , Obesity/complications , Postprandial Period/physiology , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Glucagon/metabolism , Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 1/adverse effects , Glucagon-Like Peptide 1/therapeutic use , Humans , Hypoglycemic Agents/adverse effects , Insulin , Prospective Studies , Treatment Outcome
15.
Vet J ; 242: 33-38, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30503541

ABSTRACT

The commonly used sedative α2-adrenoceptor agonist dexmedetomidine has adverse cardiovascular effects in dogs that can be prevented by concomitant administration of the peripherally acting α2-adrenoceptor antagonist MK-467. An ancillary effect of dexmedetomidine is to decrease insulin release from the pancreas, whereas MK-467 stimulates insulin release. This study assessed the effects of co-administered dexmedetomidine and MK-467 in a canine glibenclamide-induced hypoglycaemia model. In a randomised, cross-over experiment, eight beagle dogs received five intravenous treatments, comprising two administrations of saline, with dexmedetomidine or dexmedetomidine and MK-467, and three administrations of glibenclamide, with saline, dexmedetomidine or dexmedetomidine and MK-467. Plasma concentrations of glucose, lactate, insulin, glucagon and the test drugs were monitored. Administration of glibenclamide significantly increased insulin secretion and decreased blood glucose concentrations. Dexmedetomidine counteracted glibenclamide-evoked hypoglycaemia. This was opposed by the α2-adrenoceptor antagonist MK-467, but the glibenclamide-evoked hypoglycaemia was not potentiated by co-administration of dexmedetomidine and MK-467. None of the dogs developed uncontrolled hypoglycaemia. Thus, the combination of dexmedetomidine and MK-467 appeared to be safe in this canine hypoglycaemia model. Nevertheless, when MK-467 is used to alleviate the undesired cardiovascular effects of α2-adrenoceptor agonists in dogs, it should be used with caution in animals at risk for hypoglycaemia because of its insulin-releasing and hypoglycaemic effects.


Subject(s)
Adrenergic alpha-2 Receptor Agonists/pharmacology , Dexmedetomidine/pharmacology , Hypnotics and Sedatives/pharmacology , Hypoglycemia/drug therapy , Quinolizines/pharmacology , Adrenergic alpha-2 Receptor Agonists/administration & dosage , Anesthesia, Intravenous/veterinary , Animals , Blood Glucose/drug effects , Cross-Over Studies , Dexmedetomidine/administration & dosage , Disease Models, Animal , Dogs , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Glucagon/blood , Glucagon/drug effects , Glyburide , Hypnotics and Sedatives/administration & dosage , Hypoglycemia/chemically induced , Hypoglycemic Agents , Insulin/blood , Insulin/metabolism , Male , Quinolizines/administration & dosage , Random Allocation , Treatment Outcome
16.
Diabetes Obes Metab ; 19(6): 809-813, 2017 06.
Article in English | MEDLINE | ID: mdl-28128510

ABSTRACT

AIM: To examine metabolic factors that influence ketone production after sodium-glucose cotransport inhibitor (SGLT2) administration. RESEARCH DESIGN AND METHODS: Fasting plasma glucose (FPG), insulin, glucagon, free fatty acid and ketone concentrations were measured in 15 type 2 diabetes mellitus (T2DM) and 16 non-diabetic subjects before and at day 1 and day 14 after treatment with empagliflozin. RESULTS: Empagliflozin caused a 38 mg/dL reduction in FPG concentration in T2DM patients. However, it caused only a small but significant (7 mg/dL) reduction in the FPG concentration in impaired fasting glucose (IFG) subjects and did not affect FPG concentration in normal glucose tolerant (NGT) subjects. Empagliflozin caused a significant increase in mean plasma glucagon, free fatty acid (FFA) and ketone concentrations in T2DM subjects. However, empagliflozin did not cause a significant change in mean plasma insulin, glucagon or ketone concentrations in non-diabetic subjects. An index that integrates change in plasma glucose, insulin and FFA concentration at day 1 strongly correlates with plasma ketone concentration at day 1 (r = 0.85, P < .001) and day 14 (r = 0.63, r = 0.01) and predicts, with 86% sensitivity and 83% specificity, subjects at the top tertile for plasma ketone concentration after empagliflozin treatment. CONCLUSION: Results of the present study demonstrate that SGLT2 inhibition exerts different metabolic effects in non-diabetic individuals as compared to diabetic patients.


Subject(s)
Benzhydryl Compounds/administration & dosage , Diabetes Mellitus, Type 2/blood , Fasting/blood , Glucose Intolerance/blood , Glucosides/administration & dosage , Hypoglycemic Agents/administration & dosage , Ketones/blood , Sodium-Glucose Transporter 2 Inhibitors , Blood Glucose/drug effects , Diabetes Mellitus, Type 2/drug therapy , Fatty Acids, Nonesterified/blood , Female , Glucagon/blood , Glucagon/drug effects , Glucose Intolerance/drug therapy , Glucose Tolerance Test , Humans , Male , Middle Aged
17.
Diabetes ; 66(4): 960-969, 2017 04.
Article in English | MEDLINE | ID: mdl-28130310

ABSTRACT

Misregulated hormone secretion from the islet of Langerhans is central to the pathophysiology of diabetes. Although insulin plays a key role in glucose regulation, the importance of glucagon is increasingly acknowledged. However, the mechanisms that regulate glucagon secretion from α-cells are still unclear. We used pseudoislets reconstituted from dispersed islet cells to study α-cells with and without various indirect effects from other islet cells. Dispersed islet cells secrete aberrant levels of glucagon and insulin at basal and elevated glucose levels. When cultured, murine islet cells reassociate to form pseudoislets, which recover normal glucose-regulated hormone secretion, and human islet cells follow a similar pattern. We created small (∼40-µm) pseudoislets using all of the islet cells or only some of the cell types, which allowed us to characterize novel aspects of regulated hormone secretion. The recovery of regulated glucagon secretion from α-cells in small pseudoislets depends upon the combined action of paracrine factors, such as insulin and somatostatin, and juxtacrine signals between EphA4/7 on α-cells and ephrins on ß-cells. Although these signals modulate different pathways, both appear to be required for proper inhibition of glucagon secretion in response to glucose. This improved understanding of the modulation of glucagon secretion can provide novel therapeutic routes for the treatment of some individuals with diabetes.


Subject(s)
Glucagon-Secreting Cells/metabolism , Glucagon/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Actins/metabolism , Animals , Cell Communication , Cells, Cultured , Cyclic AMP/metabolism , Ephrins/metabolism , Flow Cytometry , Glucagon/drug effects , Glucagon-Secreting Cells/drug effects , Glucose/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Mice , Mice, Transgenic , Organ Culture Techniques , Paracrine Communication , Receptor, EphA4/metabolism , Receptor, EphA7/metabolism , Signal Transduction , Somatostatin/metabolism
18.
Diabetes ; 66(4): 987-993, 2017 04.
Article in English | MEDLINE | ID: mdl-28082457

ABSTRACT

Xenocell therapy from neonate or adult pig pancreatic islets is one of the most promising alternatives to allograft in type 1 diabetes for addressing organ shortage. In humans, however, natural and elicited antibodies specific for pig xenoantigens, α-(1,3)-galactose (GAL) and N-glycolylneuraminic acid (Neu5Gc), are likely to significantly contribute to xenoislet rejection. We obtained double-knockout (DKO) pigs lacking GAL and Neu5Gc. Because Neu5Gc-/- mice exhibit glycemic dysregulations and pancreatic ß-cell dysfunctions, we evaluated islet function and glucose metabolism regulation in DKO pigs. Isolation of islets from neonate piglets yielded identical islet equivalent quantities to quantities obtained from control wild-type pigs. In contrast to wild-type islets, DKO islets did not induce anti-Neu5Gc antibody when grafted in cytidine monophosphate-N-acetylneuraminic acid hydroxylase KO mice and exhibited in vitro normal insulin secretion stimulated by glucose and theophylline. Adult DKO pancreata showed no histological abnormalities, and immunostaining of insulin and glucagon was similar to that from wild-type pancreata. Blood glucose, insulin, C-peptide, the insulin-to-glucagon ratio, and HOMA-insulin resistance in fasted adult DKO pigs and blood glucose and C-peptide changes after intravenous glucose or insulin administration were similar to wild-type pigs. This first evaluation of glucose homeostasis in DKO pigs for two major xenoantigens paves the way to their use in (pre)clinical studies.


Subject(s)
Galactose/genetics , Glucose/pharmacology , Insulin/metabolism , Islets of Langerhans/drug effects , Neuraminic Acids/metabolism , Purinergic P1 Receptor Antagonists/pharmacology , Theophylline/pharmacology , Animals , Antigens, Heterophile , Blood Glucose/drug effects , Blood Glucose/metabolism , C-Peptide/drug effects , C-Peptide/metabolism , Diabetes Mellitus, Type 1/surgery , Galactose/immunology , Gene Knockout Techniques , Glucagon/drug effects , Glucagon/metabolism , Homeostasis , Insulin Secretion , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Islets of Langerhans Transplantation , Male , Neuraminic Acids/immunology , Pancreas/metabolism , Swine , Transplantation, Heterologous
19.
Am J Physiol Endocrinol Metab ; 312(3): E127-E135, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27998960

ABSTRACT

Recent epidemiological studies have revealed novel relationships between low water intake or high vasopressin (AVP) and the risk of hyperglycemia and diabetes. AVP V1A and V1B receptors (R) are expressed in the liver and pancreatic islets, respectively. The present study was designed to determine the impact of different levels of circulating AVP on glucose homeostasis in normal Sprague-Dawley rats, as well as the respective roles of V1AR and V1BR. We showed that acute injection of AVP induces a dose-dependent increase in glycemia. Pretreatment with a selective V1AR antagonist, but not a V1BR antagonist, dose-dependently prevented the rise in glycemia. V1BR antagonism did not modify the hyperinsulinemic response, resulting from AVP-induced hyperglycemia, but enhanced the fall in glucagonemia. Acute administration of selective V1AR or V1BR agonists confirmed the involvement of V1AR in the hyperglycemic effect of AVP. In chronic experiments, AVP levels were altered in both directions. Sustained AVP infusion through implantable minipumps induced a time-dependent increase in fasting glycemia, whereas lowering endogenous AVP by increasing water intake had no effect. After 4 wk of AVP infusion, the rise in glycemia amounted to 1.1 mmol/l (P < 0.01) without significant change in insulinemia. This effect was attenuated by cotreatment with a V1AR antagonist. Similar results were observed in lean Zucker rats. These findings demonstrate for the first time a causal link between chronic high AVP and hyperglycemia through V1AR activation and, thus, provide a pathophysiological explanation for the relationship observed in human cohorts between the AVP-hydration axis and the risk of diabetes.


Subject(s)
Arginine Vasopressin/pharmacology , Blood Glucose/drug effects , Glucagon/drug effects , Hyperglycemia/blood , Receptors, Vasopressin/drug effects , Animals , Antidiuretic Hormone Receptor Antagonists/pharmacology , Blood Glucose/metabolism , Gene Knock-In Techniques , Glucagon/blood , Hyperinsulinism/blood , Indoles/pharmacology , Insulin/blood , Male , Optical Imaging , Pancreas/metabolism , Peptides, Cyclic/pharmacology , Pyrrolidines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Zucker , Receptors, Vasopressin/agonists , Receptors, Vasopressin/metabolism
20.
Diabetes ; 66(2): 264-271, 2017 02.
Article in English | MEDLINE | ID: mdl-27554476

ABSTRACT

In addition to its pivotal role in psychosocial behavior, the hypothalamic neuropeptide oxytocin contributes to metabolic control by suppressing eating behavior. Its involvement in glucose homeostasis is less clear, although pilot experiments suggest that oxytocin improves glucose homeostasis. We assessed the effect of intranasal oxytocin (24 IU) administered to 29 healthy, fasted male subjects on glucose homeostasis measured by means of an oral glucose tolerance test. Parameters of glucose metabolism were analyzed according to the oral minimal model. Oxytocin attenuated the peak excursion of plasma glucose and augmented the early increases in insulin and C-peptide concentrations in response to the glucose challenge, while slightly blunting insulin and C-peptide peaks. Oral minimal model analyses revealed that oxytocin compared with placebo induced a pronounced increase in ß-cell responsivity (PHItotal) that was largely due to an enhanced dynamic response (PHId), and a more than twofold improvement in glucose tolerance (disposition index). Adrenocorticotropic hormone (ACTH), cortisol, glucagon, and nonesterified fatty acid (NEFA) concentrations were not or were only marginally affected. These results indicate that oxytocin plays a significant role in the acute regulation of glucose metabolism in healthy humans and render the oxytocin system a potential target of antidiabetic treatment.


Subject(s)
Blood Glucose/drug effects , Insulin-Secreting Cells/drug effects , Oxytocics/pharmacology , Oxytocin/pharmacology , Adrenocorticotropic Hormone/drug effects , Adrenocorticotropic Hormone/metabolism , Adult , Blood Glucose/metabolism , C-Peptide/drug effects , C-Peptide/metabolism , Fatty Acids, Nonesterified/metabolism , Glucagon/drug effects , Glucagon/metabolism , Glucose Tolerance Test , Healthy Volunteers , Humans , Hydrocortisone/metabolism , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...