Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 93
Filter
1.
Int J Mol Sci ; 22(11)2021 May 27.
Article in English | MEDLINE | ID: mdl-34072220

ABSTRACT

Insulin plays a significant role in carbohydrate homeostasis as the blood glucose lowering hormone. Glucose-induced insulin secretion (GSIS) is augmented by glucagon-like peptide (GLP-1), a gastrointestinal peptide released in response to ingesting nutriments. The secretion of insulin and GLP-1 is mediated by the binding of nutrients to G protein-coupled receptors (GPCRs) expressed by pancreatic ß-cells and enteroendocrine cells, respectively. Therefore, insulin secretagogues and incretin mimetics currently serve as antidiabetic treatments. This study demonstrates the potency of synthetic isoprenoid derivatives of lysophosphatidylcholines (LPCs) to stimulate GSIS and GLP-1 release. Murine insulinoma cell line (MIN6) and enteroendocrinal L cells (GLUTag) were incubated with LPCs bearing geranic acid (1-GA-LPC), citronellic acid (1-CA-LPC), 3,7-dimethyl-3-vinyloct-6-enoic acid (GERA-LPC), and (E)-3,7,11-trimethyl- 3-vinyldodeca-6,10-dienoic acid (1-FARA-LPC). Respective free terpene acids were also tested for comparison. Besides their insulin- and GLP-1-secreting capabilities, we also investigated the cytotoxicity of tested compounds, the ability to intracellular calcium ion mobilization, and targeted GPCRs involved in maintaining lipid and carbohydrate homeostasis. We observed the high cytotoxicity of 1-GERA-LPC and 1-FARA-LPC in contrast 1-CA-LPC and 1-GA-LPC. Moreover, 1-CA-LPC and 1-GA-LPC demonstrated the stimulatory effect on GSIS and 1-CA-LPC augmented GLP-1 secretion. Insulin and GLP-1 release appeared to be GPR40-, GPR55-, GPR119- and GPR120-dependent.


Subject(s)
Glucagon-Like Peptide 1/biosynthesis , Insulin Secretion/drug effects , Insulin/biosynthesis , Lipid Metabolism/drug effects , Lysophosphatidylcholines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Terpenes/pharmacology , Calcium/metabolism , Humans , Intracellular Space/metabolism , Lysophosphatidylcholines/chemistry , Molecular Structure , Terpenes/chemistry
2.
Cell Death Dis ; 12(1): 113, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33479193

ABSTRACT

In the status of obesity, the glucagon-like peptide-1 (GLP-1) level usually declines and results in metabolic syndrome. This study aimed to investigate the intracellular mechanism of GLP-1 synthesis in L cells from the perspective of microRNA (miRNA). In the present study, we found that GLP-1 level was down-regulated in the plasma and ileum tissues of obese mice, while the ileac miR-194 expression was up-regulated. In vitro experiments indicated that miR-194 overexpression down-regulated GLP-1 level, mRNA levels of proglucagon gene (gcg) and prohormone convertase 1/3 gene (pcsk1), and the nuclear protein level of beta-catenin (ß-catenin). Further investigation confirmed that ß-catenin could promote gcg transcription through binding to transcription factor 7-like 2 (TCF7L2). miR-194 suppressed gcg mRNA level via negatively regulating TCF7L2 expression. What's more, forkhead box a1 (Foxa1) could bind to the promoter of pcsk1 and enhanced its transcription. miR-194 suppressed pcsk1 transcription through targeting Foxa1. Besides, the interference of miR-194 reduced palmitate (PA)-induced cell apoptosis and the anti-apoptosis effect of miR-194 inhibitor was abolished by TCF7L2 knockdown. Finally, in HFD-induced obese mice, the silence of miR-194 significantly elevated GLP-1 level and improved the metabolic symptoms caused by GLP-1 deficiency. To sum up, our study found that miR-194 suppressed GLP-1 synthesis in L cells via inhibiting TCF7L2-mediated gcg transcription and Foxa1-mediated pcsk1 transcription. Meanwhile, miR-194 took part in the PA-induced apoptosis of L cells.


Subject(s)
Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/biosynthesis , MicroRNAs/metabolism , Obesity/metabolism , Animals , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , L Cells , Male , Mice , MicroRNAs/genetics , Obesity/genetics , Transfection
3.
Cytokine ; 138: 155379, 2021 02.
Article in English | MEDLINE | ID: mdl-33271384

ABSTRACT

BACKGROUND: Blood has been the usual biological fluid for measuring analytes, but there is mounting evidence that saliva may be also useful for detecting cytokines in a noninvasive way. Thus, in this study we aimed to determine concentration of cytokines and other analytes in saliva from a population of healthy children. METHODS: We collected un-stimulated whole saliva samples from clinically healthy children, and concentration of 17 cytokines and 12 other analytes were measured in supernatants. All values were adjusted by albumin content and were log-transformed before multivariate statistical analysis. RESULTS: We included 114 children (53.5% females) between 6.0 and 11.9 years old. The highest concentrations (medians, pg/µg albumin) were seen for visfatin (183.70) and adiponectin (162.26) and the lowest for IL-13 and IL-2 (~0.003). Albumin concentration was associated with age (rS = 0.39, p < 0.001). In the multivariate analysis, five analytes (C peptide, ghrelin, GLP-1, glucagon, leptin) inversely correlated with age and positively with height-for-age. Age was also positively associated with PAI-1, while height-for-age was also positively associated with insulin and visfatin. Finally, BMI-for-age had a positive correlation with GM-CSF and insulin. CONCLUSIONS: Herein, we provided concentration values for 29 analytes in saliva from healthy children that may be useful as preliminary reference framework in the clinical research setting.


Subject(s)
Cytokines/metabolism , Saliva/metabolism , Adiponectin/biosynthesis , Age Factors , Body Height , C-Peptide/biosynthesis , Child , Cytokines/biosynthesis , Female , Ghrelin/biosynthesis , Glucagon/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Humans , Insulin/metabolism , Interleukin-13/biosynthesis , Interleukin-2/biosynthesis , Leptin/biosynthesis , Male , Multivariate Analysis , Nicotinamide Phosphoribosyltransferase/biosynthesis , Reference Values
4.
Nutrients ; 12(11)2020 Oct 23.
Article in English | MEDLINE | ID: mdl-33113929

ABSTRACT

BACKGROUND: The prevalence of type 2 diabetes is on the increase worldwide, and it represents about 90% of adults who are diagnosed with diabetes. Overweight and obesity, lifestyle, genetic predisposition and gut microbiota dysbiosis have been implicated as possible risk factors in the development of type 2 diabetes. In particular, low intake of dietary fibre and consumption of foods high in fat and sugar, which are common in western lifestyle, have been reported to contribute to the depletion of specific bacterial taxa. Therefore, it is possible that intake of high dietary fibre may alter the environment in the gut and provide the needed substrate for microbial bloom. AIM: The current review is a systematic review and meta-analysis which evaluated the role of dietary fibre in modulating gut microbiota dysbiosis in patients with type 2 diabetes. METHODS: This is a systematic review and meta-analysis of randomised controlled trials which relied on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) framework. Electronic searches were conducted using EBSCOHost with links to Health Sciences Research Databases, EMBASE and Google Scholar. The reference lists of articles were also searched for relevant studies. Searches were conducted from date of commencement of the database to 5 August 2020. The search strategy was based on the Population, Intervention, Comparator, Outcomes, Studies (PICOS) framework and involved the use of synonyms and medical subject headings (MesH). Search terms were combined with Boolean operators (OR/AND). RESULTS: Nine studies which met the inclusion criteria were selected for the systematic review and meta-analysis, and four distinct areas were identified: the effect of dietary fibre on gut microbiota; the role of dietary fibre on short-chain fatty acids (SCFAs); glycaemic control; and adverse events. There was significant difference (p < 0.01) in the relative abundance of Bifidobacterium with a mean difference of 0.72 (95% CI, 0.56, 0.89) between the dietary fibre group compared with placebo. In relation to the meta-analysis for SCFAs, while there was significant difference (p = 0.02) between the dietary fibre group and placebo with a standardised mean difference of 0.5 (95% CI, 0.08, 0.91) regarding total SCFAs, the differences were not significant (p > 0.05) in relation to acetic acid, propionic acid and butyric acid. There was only significant improvement (p = 0.002) with respect to glycated haemoglobin with a mean difference of -0.18 (95% CI, -0.29, -0.06) between the dietary fibre group and placebo group. Differences between the two groups were not significant (p > 0.05) in relation to fasting blood glucose and homeostatic model assessment of insulin resistance (HOMA-IR). Furthermore, there were no significant differences between the two groups in subjects who reported adverse events. It is possible that the promotion of SCFA producers in greater diversity and abundance by dietary fibre in this review led to improvement in glycated haemoglobin, partly due to increased glucagon-like peptide-1 (GLP-1) production. In addition, Bifidobacterium lactis has been reported to increase glycogen synthesis, decrease expression of hepatic gluconeogenesis genes, improve translocation of glucose transport-4 and promote glucose uptake. It is also possible that the reduction in body weight of participants in the intervention group compared with control may have contributed to the observed improvement in glycated haemoglobin. CONCLUSION: This systematic review and meta-analysis have demonstrated that dietary fibre can significantly improve (p < 0.05) the relative abundance of Bifidobacterium, total SCFAs and glycated haemoglobin. However, dietary fibre did not appear to have significant effect (p > 0.05) on fasting blood glucose, HOMA-IR, acetic acid, propionic acid, butyric acid and adverse events.


Subject(s)
Diabetes Mellitus, Type 2/microbiology , Dietary Fiber/pharmacology , Dysbiosis/diet therapy , Gastrointestinal Microbiome/drug effects , Adult , Diabetes Mellitus, Type 2/blood , Dysbiosis/etiology , Fatty Acids, Volatile/blood , Female , Glucagon-Like Peptide 1/biosynthesis , Glycated Hemoglobin/drug effects , Glycemic Control/methods , Humans , Male , Randomized Controlled Trials as Topic
5.
Molecules ; 25(14)2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32679813

ABSTRACT

Anesthetics, particularly volatile anesthetics, have been shown to impair glucose metabolism and cause hyperglycemia, closely linking them with mortality and morbidity as related to surgery. Beyond being an anesthetic used for general anesthesia and sedation, intravenous hypnotic propofol displays an effect on glucose metabolism. To extend the scope of propofol studies, its effects on glucose metabolism were evaluated in male Sprague-Dawley rats of various ages. Unlike chloral hydrate and isoflurane, propofol had little effect on basal glucose levels in rats at 2 months of age, although it did reduce chloral hydrate- and isoflurane-induced hyperglycemia. Propofol reduced postload glucose levels after either intraperitoneal or oral administration of glucose in both 7- and 12-month-old rats, but not those at 2 months of age. These improved effects regarding propofol on glucose metabolism were accompanied by an increase in insulin, fibroblast growth factor-21 (FGF-21), and glucagon-like peptide-1 (GLP-1) secretion. Additionally, an increase in hepatic FGF-21 expression, GLP-1 signaling, and FGF-21 signaling, along with a decrease in endoplasmic reticulum (ER) stress, were noted in propofol-treated rats at 7 months of age. Current findings imply that propofol may turn into insulin-sensitizing molecules during situations of existing insulin resistance, which involve FGF-21, GLP-1, and ER stress.


Subject(s)
Blood Glucose/drug effects , Fibroblast Growth Factors/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Glucose/metabolism , Propofol/pharmacology , Animals , Disease Models, Animal , Glucose Tolerance Test , Hyperglycemia/blood , Hyperglycemia/diagnosis , Hyperglycemia/drug therapy , Hyperglycemia/etiology , Insulin/metabolism , Liver , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects
6.
Mol Metab ; 39: 101014, 2020 09.
Article in English | MEDLINE | ID: mdl-32413586

ABSTRACT

OBJECTIVES: Our study shows that glucagon-like peptide-1 (GLP-1) is secreted within human islets and may play an unexpectedly important paracrine role in islet physiology and pathophysiology. It is known that α cells within rodent and human pancreatic islets are capable of secreting GLP-1, but little is known about the functional role that islet-derived GLP-1 plays in human islets. METHODS: We used flow cytometry, immunohistochemistry, perifusions, and calcium imaging techniques to analyse GLP-1 expression and function in islets isolated from cadaveric human donors with or without type 2 diabetes. We also used immunohistochemistry to analyse GLP-1 expression within islets from pancreatic biopsies obtained from living donors. RESULTS: We have demonstrated that human islets secrete ∼50-fold more GLP-1 than murine islets and that ∼40% of the total human α cells contain GLP-1. Our results also confirm that dipeptidyl peptidase-4 (DPP4) is expressed in α cells. Sitagliptin increased GLP-1 secretion from cultured human islets but did not enhance glucose-stimulated insulin secretion (GSIS) in islets from non-diabetic (ND) or type 2 diabetic (T2D) donors, suggesting that ß cell GLP-1 receptors (GLP-1R) may already be maximally activated. Therefore, we tested the effects of exendin-9, a GLP-1R antagonist. Exendin-9 was shown to reduce GSIS by 39% and 61% in ND islets and T2D islets, respectively. We also observed significantly more GLP-1+ α cells in T2D islets compared with ND islets obtained from cadaveric donors. Furthermore, GLP-1+ α cells were also identified in pancreatic islet sections obtained from living donors undergoing surgery. CONCLUSIONS: In summary, we demonstrated that human islets secrete robust amounts of GLP-1 from an α cell subpopulation and that GLP-1R signalling may support GSIS to a greater extent in T2D islets.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Animals , Biomarkers , Diabetes Mellitus, Type 2/etiology , Gene Expression , Glucagon/metabolism , Glucagon-Like Peptide 1/genetics , Glucose/metabolism , Humans , Immunophenotyping , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Mice
7.
Int J Mol Sci ; 21(8)2020 Apr 19.
Article in English | MEDLINE | ID: mdl-32325890

ABSTRACT

Obesity is a multifactorial disease that can lead to other health issues. Glucagon-like peptide-1(GLP-1), as one of the satiety signal, has been linked with appetite suppression and weight loss. Due to the limitations of GLP-1 and its analogues, alternative treatments such as herbal therapies have become popular. The herbal formula RCM-107 has demonstrated its inhibitory effects on lipid and carbohydrate absorption in our previous work. However, no published data described its effects on GLP-1 secretion. Therefore, this study aimed to determine the effects of RCM-107 and its individual ingredients on GLP-1 secretion via enzyme-linked immunosorbent assay (ELISA). Furthermore, molecular docking was performed to predict the key chemical compounds that are likely to be GLP-1 receptor agonists. Gardeniae fructus, one of the ingredients in RCM-107, demonstrated significantly greater effects on inducing GLP-1 secretion than the positive control epigallocatechin gallate (EGCG). Two Gardeniae fructus ligands, 3-epioleanolic acid and crocin were predicted to bind to the active form of GLP-1 receptor at the binding pocket with residues known for the receptor activation, suggesting that they could potentially serve as receptor agonists. Overall, this study reported the effects of researched herbs on GLP-1 secretion and proposed two compounds that may be responsible for antiobesity via GLP-1 receptor activation.


Subject(s)
Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/chemistry , Molecular Docking Simulation , Molecular Dynamics Simulation , Weight Loss/drug effects , Binding Sites , Cell Line , Cell Survival/drug effects , Humans , Molecular Conformation , Protein Binding , Structure-Activity Relationship
8.
J Mol Endocrinol ; 63(3): 215-226, 2019 10.
Article in English | MEDLINE | ID: mdl-31426028

ABSTRACT

GLP-1 is a potent glucose-dependent insulinotropic hormone derived from intestinal L cells. Inflammatory Interleukin-27 (IL-27), a pleiotropic two-chain cytokine, is composed of EBI3 and IL-27 p28 subunits. IL-27 has a protective effect on pancreatic ß-cell function. The relationship between IL-27 and GLP-1 is still unexplored. Here we showed interleukin-27-stimulated GLP-1 production via the Stat3-mTOR-dependent mechanism. Interleukin 27 receptor subunit alpha (IL-27 Rα) was detected in ileum and STC-1 cells. Co-localization of EBI3 and GLP-1 was observed not only in mouse ileums but also in human ileums and colons. Third-ventricular infusion of IL-27 increased ileal and plasma GLP-1 in both lean C57BL/6J mice and diet-induced obese and diabetic mice. These changes were associated with a significant increase in Stat3-mTOR activity. Treatment of STC-1 cells with IL-27 contributed to the increments of Stat3-mTOR signaling and GLP-1. Interference of mTOR activity by mTOR siRNA or rapamycin abolished the stimulation of GLP-1 production induced by IL-27 in STC-1 cells. Stat3 siRNA also blocked the stimulus effect of IL-27 on GLP-1. IL-27 increased the interaction of mTOR and Stat3 in STC-1 cells. Our results identify Stat3-mTOR as a critical signaling pathway for the stimulation of GLP-1 induced by IL-27.


Subject(s)
Glucagon-Like Peptide 1/biosynthesis , Interleukin-27/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line , Humans , Interferon-gamma/pharmacology , Interleukins/metabolism , Intestinal Mucosa/metabolism , Male , Mice, Inbred C57BL , Mice, Obese , Minor Histocompatibility Antigens/metabolism , Models, Biological , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin/genetics , Receptors, Interleukin/metabolism , Sirolimus/pharmacology
9.
Am J Physiol Renal Physiol ; 317(4): F1010-F1021, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31390233

ABSTRACT

Glucagon-like peptide-1 (GLP-1), an incretin hormone, has diuretic and natriuretic effects. The present study was designed to explore the possible underlying mechanisms for the diuretic and natriuretic effects of GLP-1 via renal nerves in rats. Immunohistochemistry revealed that GLP-1 receptors were avidly expressed in the pelvic wall, the wall being adjacent to afferent renal nerves immunoreactive to calcitonin gene-related peptide, which is the dominant neurotransmitter for renal afferents. GLP-1 (3 µM) infused into the left renal pelvis increased ipsilateral afferent renal nerve activity (110.0 ± 15.6% of basal value). Intravenous infusion of GLP-1 (1 µg·kg-1·min-1) for 30 min increased renal sympathetic nerve activity (RSNA). After the distal end of the renal nerve was cut to eliminate the afferent signal, the increase in efferent renal nerve activity during intravenous infusion of GLP-1 was diminished compared with the increase in total RSNA (17.0 ± 9.0% vs. 68.1 ± 20.0% of the basal value). Diuretic and natriuretic responses to intravenous infusion of GLP-1 were enhanced by total renal denervation (T-RDN) with acute surgical cutting of the renal nerves. Selective afferent renal nerve denervation (A-RDN) was performed by bilateral perivascular application of capsaicin on the renal nerves. Similar to T-RDN, A-RDN enhanced diuretic and natriuretic responses to GLP-1. Urine flow and Na+ excretion responses to GLP-1 were not significantly different between T-RDN and A-RDN groups. These results indicate that the diuretic and natriuretic effects of GLP-1 are partly governed via activation of afferent renal nerves by GLP-1 acting on sensory nerve fibers within the pelvis of the kidney.


Subject(s)
Afferent Pathways/drug effects , Diuresis/drug effects , Glucagon-Like Peptide 1/pharmacology , Kidney/drug effects , Kidney/innervation , Natriuresis/drug effects , Animals , Calcitonin Gene-Related Peptide/physiology , Denervation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide-1 Receptor/biosynthesis , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/immunology , HEK293 Cells , Humans , Kidney Pelvis/drug effects , Kidney Pelvis/innervation , Male , Rats , Rats, Sprague-Dawley , Renal Circulation/drug effects , Sodium/urine , Sympathetic Nervous System/drug effects , Urodynamics/drug effects
10.
Am J Physiol Endocrinol Metab ; 317(3): E494-E502, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31237451

ABSTRACT

Bile acids, glucagon-like peptide-1 (GLP-1), and fibroblast growth factor 19 (FGF19) play an important role in postprandial metabolism. In this study, we investigated the postprandial bile acid response in plasma and its relation to insulin, GLP-1, and FGF19. First, we investigated the postprandial response to 40-h fast. Then we administered glycine-conjugated deoxycholic acid (gDCA) with the meal. We performed two separate observational randomized crossover studies on healthy, lean men. In experiment 1: we tested 4-h mixed meal after an overnight fast and a 40-h fast. In experiment 2, we tested a 4-h mixed meal test with and without gDCA supplementation. Both studies measured postprandial glucose, insulin, bile acids, GLP-1, and FGF19. In experiment 1, 40 h of fasting induced insulin resistance and increased postprandial GLP-1 and FGF19 concentrations. After an overnight fast, we observed strong correlations between postprandial insulin and gDCA levels at specific time points. In experiment 2, administration of gDCA increased GLP-1 levels and lowered late postprandial glucose without effect on FGF19. Energy expenditure was not affected by gDCA administration. Unexpectedly, 40 h of fasting increased both GLP-1 and FGF19, where the former appeared bile acid independent and the latter bile acid dependent. Second, a single dose of gDCA increased postprandial GLP-1. Therefore, our data add complexity to the physiological regulation of the enterokines GLP-1 and FGF19 by bile acids.


Subject(s)
Bile Acids and Salts/pharmacology , Fasting/physiology , Fibroblast Growth Factors/biosynthesis , Glucagon-Like Peptide 1/biosynthesis , Bile Acids and Salts/blood , Blood Glucose , Cross-Over Studies , Deoxycholic Acid/pharmacology , Dietary Supplements , Energy Metabolism , Fibroblast Growth Factors/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin/blood , Insulin Resistance , Male , Postprandial Period , Young Adult
11.
Mol Biol Rep ; 46(5): 4799-4808, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31228040

ABSTRACT

Maintenance of glucose homeostasis is reciprocally regulated by insulin and glucagon-like peptide-1 (GLP-1). We previously reported that GLP-1 secretion in response to an oral glucose load was impaired following an administration of 3-deoxyglucosone (3DG), an independent factor associated with the development of pre-diabetes. Here we investigated the effects of 3DG on insulin signaling and insulin-induced GLP-1 secretion under high-glucose conditions in the enteroendocrine L cell line STC-1. STC-1 cells were exposed to 3DG (80, 300, and 1000 ng/ml) in the presence of 10-7 M insulin and 25 mM glucose. GLP-1 secretion was determined by ELISA, glucose uptake was monitored with 2-NBDG (2-(N(7-nitrobenz-2-oxa-1,3-diazol-4-yl) amino)-2-deoxyglucose), glucose consumption was detected by glucoseoxidase, and protein expression of insulin signaling molecules was examined by western blot. Results showed a decrease in insulin-induced GLP-1 secretion and insulin receptor phosphorylation after 3DG treatment. Concomitantly, 3DG treatment inhibited insulin-induced phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt) pathway activation. In the presence, but not absence, of insulin, 3DG treatment decreased insulin-stimulated glucose consumption. Inhibition of PI3K with Wortmannin attenuated insulin-induced increment in glucose transporter 2 (GLUT2) expression and 2-NBDG uptake. Accordingly, insulin-induced increase in GLUT2 expression and 2-NBGD uptake was significantly inhibited by 3DG treatment. 3DG-mediated reduction in GLUT2 expression contributes to the attenuation of insulin-induced GLP-1 secretion under high-glucose conditions in part through the insulin-PI3K/Akt/GLUT2 pathway in STC-1 cells. We conclude that 3DG interferes with insulin signaling and attenuates insulin action on glucose-induced GLP-1 secretion in STC-1 cells.


Subject(s)
Deoxyglucose/analogs & derivatives , Enteroendocrine Cells/metabolism , Glucagon-Like Peptide 1/biosynthesis , Glucose/metabolism , Insulin/metabolism , Signal Transduction , Animals , Cell Line , Deoxyglucose/metabolism , Glucose Transporter Type 2/metabolism , Mice , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/metabolism
12.
J Biosci ; 43(5): 921-929, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30541952

ABSTRACT

5rolGLP-HV is a promising dual-function peptide for the treatment of diabetes and thrombosis simultaneously. For investigating the therapeutic mechanism of 5rolGLP-HV for type 2 diabetes mellitus (T2DM), STZ-induced diabetic mice were established and treated with 5rolGLP-HV. The results showed that daily water and food intake, blood glucose, serum and pancreatic insulin levels significantly decreased after 5rolGLP-HV treatment with various oral concentrations, and 16 mg/kg was the optimal dose for controlling diabetes. 5rolGLP-HV treatment decreased the MDA levels and the T-SOD activity in serum and pancreatic of diabetic mice (but not up to significant difference), and significantly increased the expression of signal pathways related genes of rolGLP-1, also the density of insulin expression and the numbers of apoptosis cells in islets of diabetic mice were significantly decreased in comparison to the negative diabetic mice. These effects above may be clarified the hypoglycemic mechanisms of 5rolGLP-HV, and 5rolGLP-HV may be as a potential drug for diabetes in future.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Glucagon-Like Peptide 1/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/blood , Recombinant Proteins/pharmacology , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Drinking/drug effects , Drug Evaluation, Preclinical , Eating/drug effects , Gene Expression Regulation , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide-1 Receptor/genetics , Glucagon-Like Peptide-1 Receptor/metabolism , Hirudins/chemistry , Hypoglycemic Agents/metabolism , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , Male , Malondialdehyde/antagonists & inhibitors , Malondialdehyde/metabolism , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/biosynthesis , Streptozocin , Superoxide Dismutase/metabolism
13.
EBioMedicine ; 32: 201-214, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29859856

ABSTRACT

BACKGROUND: The mechanism by which Roux-en-Y Gastric Bypass (RYGB) increases the secretion of glucagon-like peptide-1 (GLP-1) remains incompletely defined. Here we investigated whether TGR5-mTORC1 signaling mediates the RYGB-induced alteration in GLP-1 production in mice and human beings. METHODS: Circulating bile acids, TGR5-mTORC1 signaling, GLP-1 synthesis and secretion were determined in lean or obese male C57BL/6 mice with or without RYGB operation, as well as in normal glycemic subjects, obese patients with type 2 diabetes before and after RYGB. RESULTS: Positive relationships were observed among circulating bile acids, ileal mechanistic target of rapamycin complex 1 (mTORC1) signaling and GLP-1 during changes in energy status in the present study. RYGB increased circulating bile acids, ileal Takeda G protein-coupled receptor 5 (TGR5) and mTORC1 signaling activity, as well as GLP-1 production in both mice and human subjects. Inhibition of ileal mTORC1 signaling by rapamycin significantly attenuated the stimulation of bile acid secretion, TGR5 expression and GLP-1 synthesis induced by RYGB in lean and diet-induced obese mice. GLP-1 production and ileal TGR5-mTORC1 signaling were positively correlated with plasma deoxycholic acid (DCA) in mice. Treatment of STC-1 cells with DCA stimulated the production of GLP-1. This effect was associated with a significant enhancement of TGR5-mTORC1 signaling. siRNA knockdown of mTORC1 or TGR5 abolished the enhancement of GLP-1 synthesis induced by DCA. DCA increased interaction between mTOR-regulatory-associated protein of mechanistic target of rapamycin (Raptor) and TGR5 in STC-1 cells. INTERPRETATION: Deoxycholic acid-TGR5-mTORC1 signaling contributes to the up-regulation of GLP-1 production after RYGB.


Subject(s)
Diabetes Mellitus, Type 2/surgery , Gastric Bypass/adverse effects , Glucagon-Like Peptide 1/genetics , Obesity/surgery , Receptors, G-Protein-Coupled/genetics , Animals , Bile Acids and Salts/blood , Blood Glucose , Deoxycholic Acid/blood , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation/genetics , Glucagon-Like Peptide 1/biosynthesis , Humans , Insulin Resistance/genetics , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mice , Mice, Obese , Middle Aged , Obesity/blood , Obesity/genetics , Obesity/pathology , Signal Transduction/genetics
14.
Peptides ; 100: 68-74, 2018 02.
Article in English | MEDLINE | ID: mdl-29412834

ABSTRACT

A new strategy under development for the treatment of type 2 diabetes and obesity is to mimic some of the effects of bariatric surgery by delivering food-related stimuli to the distal gastrointestinal tract where they should enhance the release of gut hormones such as glucagon-like peptide-1 (GLP-1) and peptideYY (PYY). Methods include inhibition of food digestion and absorption in the upper GI tract, or oral delivery of stimuli in capsules or pelleted form to protect them against gastric degradation. A variety of agents have been tested in humans using capsules, microcapsules or pellets, delivering nutrients, bile acids, fatty acids and bitter compounds. This review examines the outcomes of these different approaches and supporting evidence from intestinal perfusion studies.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gastrointestinal Tract/drug effects , Incretins/therapeutic use , Obesity/drug therapy , Bariatric Surgery , Diabetes Mellitus, Type 2/metabolism , Food-Drug Interactions , Gastrointestinal Tract/metabolism , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/drug effects , Humans , Incretins/chemistry , Obesity/metabolism , Peptide YY/drug effects , Peptide YY/metabolism , Secretagogues
15.
Gut ; 67(2): 271-283, 2018 02.
Article in English | MEDLINE | ID: mdl-28377388

ABSTRACT

OBJECTIVE: To investigate the beneficial role of prebiotics on endothelial dysfunction, an early key marker of cardiovascular diseases, in an original mouse model linking steatosis and endothelial dysfunction. DESIGN: We examined the contribution of the gut microbiota to vascular dysfunction observed in apolipoprotein E knockout (Apoe-/-) mice fed an n-3 polyunsaturated fatty acid (PUFA)-depleted diet for 12 weeks with or without inulin-type fructans (ITFs) supplementation for the last 15 days. Mesenteric and carotid arteries were isolated to evaluate endothelium-dependent relaxation ex vivo. Caecal microbiota composition (Illumina Sequencing of the 16S rRNA gene) and key pathways/mediators involved in the control of vascular function, including bile acid (BA) profiling, gut and liver key gene expression, nitric oxide and gut hormones production were also assessed. RESULTS: ITF supplementation totally reverses endothelial dysfunction in mesenteric and carotid arteries of n-3 PUFA-depleted Apoe-/- mice via activation of the nitric oxide (NO) synthase/NO pathway. Gut microbiota changes induced by prebiotic treatment consist in increased NO-producing bacteria, replenishment of abundance in Akkermansia and decreased abundance in bacterial taxa involved in secondary BA synthesis. Changes in gut and liver gene expression also occur upon ITFs suggesting increased glucagon-like peptide 1 production and BA turnover as drivers of endothelium function preservation. CONCLUSIONS: We demonstrate for the first time that ITF improve endothelial dysfunction, implicating a short-term adaptation of both gut microbiota and key gut peptides. If confirmed in humans, prebiotics could be proposed as a novel approach in the prevention of metabolic disorders-related cardiovascular diseases.


Subject(s)
Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Fructans/pharmacology , Gastrointestinal Microbiome/drug effects , Prebiotics , Aminopeptidases/genetics , Animals , Antimicrobial Cationic Peptides/genetics , Bacteria/drug effects , Bile Acids and Salts/biosynthesis , Bile Acids and Salts/blood , Carotid Arteries/physiology , Cecum/microbiology , Dietary Supplements , Disease Models, Animal , Fatty Acids, Omega-3/administration & dosage , Fatty Acids, Omega-3/deficiency , Gene Expression/drug effects , Glucagon-Like Peptide 1/biosynthesis , Male , Mesenteric Arteries/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , Neurotensin/genetics , Nitric Oxide/metabolism , Nitric Oxide Synthase/metabolism , Organic Anion Transporters, Sodium-Dependent/genetics , Proglucagon/genetics , Symporters/genetics , Vasodilation
16.
Peptides ; 91: 33-39, 2017 05.
Article in English | MEDLINE | ID: mdl-28300673

ABSTRACT

Some modified glucagon-like-peptide-1 (GLP-1) analogs are highly important for treating type 2 diabetes. Here we investigated whether GLP-1 analogs expressed in Lactococcus lactis could be substrates for modification and export by the nisin dehydratase and transporter enzyme. Subsequently we introduced a lysinoalanine by coupling a formed dehydroalanine with a lysine and investigated the structure and activity of the formed lysinoalanine-bridged GLP-1 analog. Our data show: (i) GLP-1 fused to the nisin leader peptide is very well exported via the nisin transporter NisT, (ii) production of leader-GLP-1 via NisT is higher than via the SEC system, (iii) leader-GLP-1 exported via NisT was more efficiently dehydrated by the nisin dehydratase NisB than when exported via the SEC system, (iv) individual serines and threonines in GLP-1 are dehydrated by NisB to a significantly different extent, (v) an introduced Ser30 is well dehydrated and can be coupled to Lys34 to form a lysinoalanine-bridged GLP-1 analog, (vi) a lysinoalanine(30-34) variant's conformation shifts in the presence of 25% trifluoroethanol towards a higher alpha helix content than observed for wild type GLP-1 under identical condition, (vii) a lysinoalanine(30-34) GLP-1 variant has retained significant activity. Taken together the data extend knowledge on the substrate specificities of NisT and NisB and their combined activity relative to export via the Sec system, and demonstrate that introducing a lysinoalanine bridge is an option for modifying therapeutic peptides.


Subject(s)
Bacterial Proteins/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/metabolism , Lactococcus lactis/metabolism , Lysinoalanine/chemistry , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Alanine/analogs & derivatives , Alanine/chemistry , Amino Acid Sequence , Bacterial Proteins/genetics , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Like Peptide 1/genetics , Hydro-Lyases/chemistry , Lactococcus lactis/genetics , Membrane Proteins/genetics , Membrane Transport Proteins/genetics , Protein Processing, Post-Translational , SEC Translocation Channels/metabolism , Serine/chemistry , Substrate Specificity , Threonine/chemistry
17.
Cell Microbiol ; 19(2)2017 02.
Article in English | MEDLINE | ID: mdl-27405092

ABSTRACT

The intestinal epithelium is an active barrier separating the host from its microbiota. It senses microbial compounds through expression of a wide range of receptors including the Toll-like receptors (TLRs). TLRs have been shown to regulate epithelium permeability or secretion of defensin by Paneth cells. However, the expression and function of TLRs in enteroendocrine L-cells, a specific subtype of intestinal cells secreting PYY and GLP-1, have not yet been assessed. PYY and GLP-1 are implicated in regulation of gut motility, food intake and insulin secretion, and are of great interest regarding obesity and type 2 diabetes. Using a cellular model of human L-cells and a reporter system for NF-κB activation pathway, we reported functional expression of TLRs in these cells. Stimulation with specific TLR-agonists increased expression of Pyy but not Proglucagon in an NF-κB-dependent manner. Moreover, the effect of TLR stimulation was additive to butyrate, a product of bacterial fermentation, on Pyy expression. Additionally, butyrate also increased Tlr expression, including Tlr4, and the NF-κB response to TLR stimulation. Altogether, our results demonstrated a role of TLRs in the modulation of Pyy expression and the importance of butyrate, a product of bacterial fermentation in regulation of microbial TLR-dependent sensing.


Subject(s)
Butyrates/metabolism , Enteroendocrine Cells/metabolism , Gene Expression Regulation , Peptide YY/metabolism , Toll-Like Receptors/agonists , Cell Line , Glucagon-Like Peptide 1/biosynthesis , Humans , NF-kappa B/metabolism
18.
Cardiovasc Res ; 113(2): 183-194, 2017 02.
Article in English | MEDLINE | ID: mdl-27702762

ABSTRACT

AIMS: Enhancement of glucagon-like peptide-1 (GLP-1) reduces glucose levels and preserves pancreatic ß-cell function, but its effect against restenosis is unknown. METHODS AND RESULTS: We investigated the effect of subcutaneous injection of exenatide or local delivery of a recombinant adenovirus expressing GLP-1 (rAd-GLP-1) into carotid artery, in reducing the occurrence of restenosis following balloon injury. As a control, we inserted ß-galactosidase cDNA in the same vector (rAd-ßGAL). Otsuka Long-Evans Tokushima rats were assigned to three groups (n = 12 each): (1) normal saline plus rAd-ßGAL delivery (NS + rAd-ßGAL), (2) exenatide plus rAd-ßGAL delivery (Exenatide + rAd-ßGAL), and (3) normal saline plus rAd-GLP-1 delivery (NS + rAd-GLP-1). Normal saline or exenatide were administered subcutaneously from 1 week before to 2 weeks after carotid injury. After 3 weeks, the NS + rAd-ßGAL group showed the highest intima-media ratio (IMR; 3.73 ± 0.90), the exenatide + rAd-ßGAL treatment was the next highest (2.80 ± 0.51), and NS + rAd-GLP-1 treatment showed the lowest IMR (1.58 ± 0.48, P < 0.05 vs. others). The proliferation and migration of vascular smooth muscle cells and monocyte adhesion were decreased significantly after rAd-GLP-1 treatment, showing the same overall patterns as the IMR. In injured vessels, the apoptosis was greater and MMP2 expression was less in the NS + rAd-GLP-1 than in the exenatide or rAd-ßGAL groups. In vitro expressions of matrix metalloproteinases-2 and monocyte chemoattractant protein-1 and nuclear factor-kappa-B-p65 translocation were decreased more in the NS + rAd-GLP-1 group than in the other two groups (all P < 0.05). CONCLUSION: Direct GLP-1 overexpression showed better protection against restenosis after balloon injury via suppression of vascular smooth muscle cell migration, increased apoptosis, and decreased inflammatory processes than systemic exenatide treatment. This has potential therapeutic implications for treating macrovascular complications in diabetes.


Subject(s)
Adenoviridae/genetics , Carotid Artery Injuries/therapy , Carotid Artery, External/metabolism , Coronary Stenosis/prevention & control , Diabetes Mellitus/therapy , Gene Transfer Techniques , Genetic Therapy , Genetic Vectors , Glucagon-Like Peptide 1/biosynthesis , Human Umbilical Vein Endothelial Cells/drug effects , Neointima , Animals , Apoptosis , Carotid Artery Injuries/genetics , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Carotid Artery, External/pathology , Cell Adhesion , Cell Movement , Cell Proliferation , Cells, Cultured , Coronary Stenosis/genetics , Coronary Stenosis/metabolism , Coronary Stenosis/pathology , Diabetes Mellitus/genetics , Diabetes Mellitus/metabolism , Disease Models, Animal , Exenatide , Glucagon-Like Peptide 1/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Hypoglycemic Agents/administration & dosage , Incretins/administration & dosage , Male , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Peptides/administration & dosage , Rats, Inbred OLETF , Transcription Factor RelA/metabolism , Transfection , Venoms/administration & dosage
19.
Benef Microbes ; 8(2): 243-255, 2017 Apr 26.
Article in English | MEDLINE | ID: mdl-28008783

ABSTRACT

Restoration of dysbiosed gut microbiota through probiotic may have profound effect on type 2 diabetes. In the present study, rats were fed high fat diet (HFD) for 3 weeks and injected with low dose streptozotocin to induce type 2 diabetes. Diabetic rats were then fed Lactobacillus rhamnosus NCDC 17 and L. rhamnosus GG with HFD for six weeks. L. rhamnosus NCDC 17 improved oral glucose tolerance test, biochemical parameters (fasting blood glucose, plasma insulin, glycosylated haemoglobin, free fatty acids, triglycerides, total cholesterol, low-density lipoprotein cholesterol and high-density lipoprotein cholesterol), oxidative stress (thiobarbituric acid reactive substance and activities of catalase, superoxide dismutase and glutathione peroxidase in blood and liver), bifidobacteria and lactobacilli in cecum, expression of glucagon like peptide-1 producing genes in cecum, and adiponection in epididymal fat, while decreased propionate proportions (%) in caecum, and expression of tumour necrosis factor-α and interlukin-6 in epididymal fat of diabetic rats as compared to diabetes control group. These findings offered a base for the use of L. rhamnosus NCDC 17 for the improvement and early treatment of type 2 diabetes.


Subject(s)
Cultured Milk Products/microbiology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Lacticaseibacillus rhamnosus , Probiotics/therapeutic use , Adiponectin/biosynthesis , Animals , Blood Glucose/metabolism , Cecum/microbiology , Diabetes Mellitus, Type 2/chemically induced , Diet, High-Fat , Dietary Supplements/microbiology , Gastrointestinal Microbiome/drug effects , Glucagon-Like Peptide 1/biosynthesis , Glucose Tolerance Test , Interleukin-6/biosynthesis , Male , Oxidative Stress , Propionates/metabolism , Rats , Rats, Wistar , Streptozocin , Tumor Necrosis Factor-alpha/biosynthesis
20.
J Diabetes Investig ; 7 Suppl 1: 50-5, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27186356

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a product of proglucagon cleavage synthesized in L cells in the intestinal mucosa, α-cells in the pancreatic islet, and neurons in the nucleus of the solitary tract. GLP-1 is essential for normal glucose tolerance and acts through a specific GLP-1 receptor that is expressed by islet ß-cells as well as other cell types. Because plasma concentrations of GLP-1 increase following meal ingestion it has been generally presumed that GLP-1 acts as a hormone, communicating information from the intestine to the endocrine pancreas through the circulation. However, there are a number of problems with this model including low circulating concentrations of GLP-1 in plasma, limited changes after meal ingestion and rapid metabolism in the plasma. Moreover, antagonism of systemic GLP-1 action impairs insulin secretion in the fasting state, suggesting that the GLP-1r is active even when plasma GLP-1 levels are low and unchanging. Consistent with these observations, deletion of the GLP-1r from islet ß-cells causes intolerance after IP or IV glucose, challenges that do not induce GLP-1 secretion. Taken together, these data support a model whereby GLP-1 acts through neural or paracrine mechanisms to regulate physiologic insulin secretion. In contrast, bariatric surgery seems to be a condition in which circulating GLP-1 could have an endocrine effect. Both gastric bypass and sleeve gastrectomy are associated with substantial increases in postprandial GLP-1 release and in these conditions interference with GLP-1r signaling has a significant impact on glucose regulation after eating. Thus, with either bariatric surgery or treatment with long-acting GLP-1r agonists, circulating peptide mediates insulinotropic activity. Overall, a case can be made that physiologic actions of GLP-1 are not hormonal, but that an endocrine mechanism of GLP-1r activation can be co-opted for therapeutics.


Subject(s)
Glucagon-Like Peptide 1/physiology , Glucagon-Like Peptide-1 Receptor/physiology , Insulin/metabolism , Animals , Fasting , Glucagon-Like Peptide 1/biosynthesis , Glucagon-Secreting Cells/metabolism , Humans , Incretins/physiology , Insulin Secretion , Proglucagon/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...