Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 102
Filter
1.
Clin Pharmacol Ther ; 111(1): 218-226, 2022 01.
Article in English | MEDLINE | ID: mdl-34312836

ABSTRACT

Serious hypoglycemia is a major adverse event associated with insulin secretagogues. Previous studies have suggested a potential relationship between angiotensin-converting enzyme inhibitors (ACEIs) used with sulfonylureas and serious hypoglycemia, and widely used drug compendia warn of this potential drug-drug interaction. We investigated the association between serious hypoglycemia and concomitant use of ACEIs in patients receiving insulin secretagogues, using the self-controlled case series design and Medicaid claims data from 5 US states linked to Medicare claims from 1999-2011. The exposure of interest was active prescription for ACEIs during insulin secretagogue or metformin (negative control object drug) episodes. The outcome was hospital presentation for serious hypoglycemia, identified by discharge diagnosis codes in inpatient and emergency department claims (positive predictive value ~ 78-89%). We calculated confounder-adjusted rate ratios (RRs) and 95% confidence internals (CIs) of outcome occurrence during ACEI-exposed vs. ACEI-unexposed time using conditional Poisson regression. The RRs for ACEIs were not statistically elevated during observation time of glipizide (RR, 1.06; CI, 0.98-1.15), glyburide (RR, 1.05; CI, 0.96-1.15), repaglinide (RR, 1.15; CI, 0.94-1.41), or metformin (RR, 1.02; CI, 0.97-1.06); but was modestly elevated with glimepiride (RR, 1.23; CI, 1.11-1.37) and modestly reduced with nateglinide (RR, 0.73; CI, 0.56-0.96). The overall pattern of results do not suggest that ACEIs used with insulin secretagogues were associated with increased rates of serious hypoglycemia, with the possible exception of glimepiride.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/adverse effects , Hypoglycemia/chemically induced , Hypoglycemia/epidemiology , Insulin/agonists , Secretagogues/adverse effects , Administrative Claims, Healthcare , Aged , Aged, 80 and over , Carbamates/adverse effects , Databases, Factual , Diabetes Mellitus, Type 2/drug therapy , Drug Interactions , Female , Glipizide/adverse effects , Glyburide/adverse effects , Humans , Hypoglycemic Agents/adverse effects , Male , Medicaid , Metformin/adverse effects , Middle Aged , Nateglinide/adverse effects , Pharmacoepidemiology , Piperidines/adverse effects , Sulfonylurea Compounds/adverse effects , United States
2.
Molecules ; 26(15)2021 Jul 30.
Article in English | MEDLINE | ID: mdl-34361788

ABSTRACT

This research investigated a UPLC-QTOF/ESI-MS-based phytochemical profiling of Combretum indicum leaf extract (CILEx), and explored its in vitro antioxidant and in vivo antidiabetic effects in a Long-Evans rat model. After a one-week intervention, the animals' blood glucose, lipid profile, and pancreatic architectures were evaluated. UPLC-QTOF/ESI-MS fragmentation of CILEx and its eight docking-guided compounds were further dissected to evaluate their roles using bioinformatics-based network pharmacological tools. Results showed a very promising antioxidative effect of CILEx. Both doses of CILEx were found to significantly (p < 0.05) reduce blood glucose, low-density lipoprotein (LDL), and total cholesterol (TC), and increase high-density lipoprotein (HDL). Pancreatic tissue architectures were much improved compared to the diabetic control group. A computational approach revealed that schizonepetoside E, melianol, leucodelphinidin, and arbutin were highly suitable for further therapeutic assessment. Arbutin, in a Gene Ontology and PPI network study, evolved as the most prospective constituent for 203 target proteins of 48 KEGG pathways regulating immune modulation and insulin secretion to control diabetes. The fragmentation mechanisms of the compounds are consistent with the obtained effects for CILEx. Results show that the natural compounds from CILEx could exert potential antidiabetic effects through in vivo and computational study.


Subject(s)
Antioxidants/pharmacology , Combretum/chemistry , Diabetes Mellitus, Experimental/drug therapy , Hypoglycemic Agents/pharmacology , Oxidative Stress/drug effects , Animals , Antioxidants/chemistry , Antioxidants/isolation & purification , Arbutin/chemistry , Arbutin/isolation & purification , Binding Sites , Blood Glucose/drug effects , Cholesterol, HDL/agonists , Cholesterol, HDL/blood , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/blood , Computational Biology/methods , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Flavonoids/chemistry , Flavonoids/isolation & purification , Gene Expression Profiling , Gene Expression Regulation , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/isolation & purification , Insulin/agonists , Insulin/metabolism , Male , Models, Molecular , Pancreas/drug effects , Pancreas/metabolism , Pancreas/pathology , Plant Extracts/chemistry , Plant Leaves/chemistry , Protein Binding , Protein Conformation , Rats , Rats, Long-Evans , Triterpenes/chemistry , Triterpenes/isolation & purification
3.
Islets ; 11(3): 51-64, 2019.
Article in English | MEDLINE | ID: mdl-31084524

ABSTRACT

The aim of the study was to determine the acute contribution of fuel oxidation in mediating the increase in insulin secretion rate (ISR) in response to fatty acids. Measures of mitochondrial metabolism, as reflected by oxygen consumption rate (OCR) and cytochrome c reduction, calcium signaling, and ISR by rat islets were used to evaluate processes stimulated by acute exposure to palmitic acid (PA). The contribution of mitochondrial oxidation of PA was determined in the presence and absence of a blocker of mitochondrial transport of fatty acids (etomoxir) at different glucose concentrations. Subsequent to increasing glucose from 3 to 20 mM, PA caused small increases in OCR and cytosolic calcium (about 20% of the effect of glucose). In contrast, the effect of PA on ISR was almost 3 times that by glucose, suggesting that the metabolism of PA is not the dominant mechanism mediating PA's effect on ISR. This was further supported by lack of inhibition of PA-stimulated OCR and ISR when blocking entry of PA into mitochondria (with etomoxir), and PA's lack of stimulation of reduced cytochrome c in the presence of high glucose. Consistent with the lack of metabolic stimulation by PA, an inhibitor of calcium release from the endoplasmic reticulum, but not a blocker of L-type calcium channels, abolished the PA-induced elevation of cytosolic calcium. Notably, ISR was unaffected by thapsigargin showing the dissociation of endoplasmic reticulum calcium release and second phase insulin secretion. In conclusion, stimulation of ISR by PA was mediated by mechanisms largely independent of the oxidation of the fuel.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Palmitates/metabolism , Animals , Endoplasmic Reticulum/drug effects , Insulin/agonists , Islets of Langerhans/drug effects , Male , Mitochondria/drug effects , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Oxygen Consumption/drug effects , Palmitates/pharmacology , Rats , Rats, Sprague-Dawley
4.
Bull Exp Biol Med ; 165(5): 644-648, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30225696

ABSTRACT

The effects of the pegylated form of glucagon-like peptide 1 (pegGLP-1) on oligopotent ß cell precursors (CD45-TER119-CD133+CD49flow) in the pancreas were studied in C57Bl/6 mice. Under conditions of streptozotocin-induced type 1 diabetes mellitus, intraperitoneal injection of pegGLP1 increased the content of ß cell precursors and dithizone-stained cells in the pancreas. ß Cell precursors of mice with diabetes demonstrated high self-maintenance potential. In contrast to pegGLP-1, native GLP-1 did not affect ß cell precursors in diabetic animals. Treatment of a culture of ß cell precursors from mice with diabetes induced the yield of dithizone-stained mononuclears. In conditioned mediums of dithizone-positive cells obtained as a result of differentiation of ß cell precursors from mice with diabetes, insulin was detected after administration of pegGLP-1 (10-7 M) and glucose (3 mmol/liter); the level of insulin increased with increasing glucose concentration (to 20 mmol/liter). The in vitro effect of pegGLP-1 did not differ from the effect of GLP-1 (10-7 M).


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Glucagon-Like Peptide 1/pharmacology , Incretins/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/agonists , Polyethylene Glycols/chemistry , Animals , Antigens, CD/metabolism , Biomarkers/metabolism , Cell Differentiation/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Glucagon-Like Peptide 1/analogs & derivatives , Incretins/chemistry , Injections, Intraperitoneal , Insulin/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Regeneration , Streptozocin
5.
J Biol Chem ; 293(43): 16818-16829, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30213860

ABSTRACT

Insulin and insulin-like growth factor 1 (IGF-1) are closely related hormones involved in the regulation of metabolism and growth. They elicit their functions through activation of tyrosine kinase-type receptors: insulin receptors (IR-A and IR-B) and IGF-1 receptor (IGF-1R). Despite similarity in primary and three-dimensional structures, insulin and IGF-1 bind the noncognate receptor with substantially reduced affinity. We prepared [d-HisB24, GlyB31, TyrB32]-insulin, which binds all three receptors with high affinity (251 or 338% binding affinity to IR-A respectively to IR-B relative to insulin and 12.4% binding affinity to IGF-1R relative to IGF-1). We prepared other modified insulins with the aim of explaining the versatility of [d-HisB24, GlyB31, TyrB32]-insulin. Through structural, activity, and kinetic studies of these insulin analogs, we concluded that the ability of [d-HisB24, GlyB31, TyrB32]-insulin to stimulate all three receptors is provided by structural changes caused by a reversed chirality at the B24 combined with the extension of the C terminus of the B chain by two extra residues. We assume that the structural changes allow the directing of the B chain C terminus to some extra interactions with the receptors. These unusual interactions lead to a decrease of dissociation rate from the IR and conversely enable easier association with IGF-1R. All of the structural changes were made at the hormones' Site 1, which is thought to interact with the Site 1 of the receptors. The results of the study suggest that merely modifications of Site 1 of the hormone are sufficient to change the receptor specificity of insulin.


Subject(s)
Insulin/agonists , Insulin/metabolism , Receptor, Insulin/metabolism , Receptors, Somatomedin/metabolism , Amino Acid Sequence , Crystallography, X-Ray , Humans , Insulin-Like Growth Factor I/chemistry , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Kinetics , Protein Binding , Receptor, IGF Type 1 , Receptor, Insulin/chemistry , Receptor, Insulin/genetics , Receptors, Somatomedin/chemistry , Receptors, Somatomedin/genetics
6.
J Autoimmun ; 94: 7-15, 2018 11.
Article in English | MEDLINE | ID: mdl-30115527

ABSTRACT

IFNα is a cytokine essential to a vast array of immunologic processes. Its induction early in the innate immune response provides a priming mechanism that orchestrates numerous subsequent pathways in innate and adaptive immunity. Despite its beneficial effects in viral infections IFNα has been reported to be associated with several autoimmune diseases including autoimmune thyroid disease, systemic lupus erythematosus, rheumatoid arthritis, primary biliary cholangitis, and recently emerged as a major cytokine that triggers Type 1 Diabetes. In this review, we dissect the role of IFNα in T1D, focusing on the potential pathophysiological mechanisms involved. Evidence from human and mouse studies indicates that IFNα plays a key role in enhancing islet expression of HLA-I in patients with T1D, thereby increasing autoantigen presentation and beta cell activation of autoreactive cytotoxic CD8 T-lymphocytes. The binding of IFNα to its receptor induces the secretion of chemokines, attracting monocytes, T lymphocytes, and NK cells to the infected tissue triggering autoimmunity in susceptible individuals. Furthermore, IFNα impairs insulin production through the induction of endoplasmic reticulum stress as well as by impairing mitochondrial function. Due to its central role in the early phases of beta cell death, targeting IFNα and its pathways in genetically predisposed individuals may represent a potential novel therapeutic strategy in the very early stages of T1D.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Diabetes Mellitus, Type 1/immunology , Gene Expression Regulation/immunology , Insulin-Secreting Cells/immunology , Interferon-alpha/immunology , Receptor, Interferon alpha-beta/immunology , Animals , Autoantigens/immunology , Autoimmunity/drug effects , Chemokines/genetics , Chemokines/immunology , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Humans , Insulin/agonists , Insulin/biosynthesis , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Interferon-alpha/antagonists & inhibitors , Interferon-alpha/genetics , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Mice , Mitochondria/drug effects , Mitochondria/immunology , Molecular Targeted Therapy/methods , Receptor, Interferon alpha-beta/antagonists & inhibitors , Receptor, Interferon alpha-beta/genetics , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology
7.
Diabetes ; 67(6): 1182-1189, 2018 06.
Article in English | MEDLINE | ID: mdl-29602791

ABSTRACT

The decrement in plasma glucose concentration with SGLT2 inhibitors (SGLT2i) is blunted by a rise in endogenous glucose production (EGP). We investigated the ability of incretin treatment to offset the EGP increase. Subjects with type 2 diabetes (n = 36) were randomized to 1) canagliflozin (CANA), 2) liraglutide (LIRA), or 3) CANA plus LIRA (CANA/LIRA). EGP was measured with [3-3H]glucose with or without drugs for 360 min. In the pretreatment studies, EGP was comparable and decreased (2.2 ± 0.1 to 1.7 ± 0.2 mg/kg ⋅ min) during a 300- to 360-min period (P < 0.01). The decrement in EGP was attenuated with CANA (2.1 ± 0.1 to 1.9 ± 0.1 mg/kg ⋅ min) and CANA/LIRA (2.2 ± 0.1 to 2.0 ± 0.1 mg/kg ⋅ min), whereas with LIRA it was the same (2.4 ± 0.2 to 1.8 ± 0.2 mg/kg ⋅ min) (all P < 0.05 vs. baseline). After CANA, the fasting plasma insulin concentration decreased (18 ± 2 to 12 ± 2 µU/mL, P < 0.05), while it remained unchanged in LIRA (18 ± 2 vs. 16 ± 2 µU/mL) and CANA/LIRA (17 ± 1 vs. 15 ± 2 µU/mL). Mean plasma glucagon did not change during the pretreatment studies from 0 to 360 min, while it increased with CANA (69 ± 3 to 78 ± 2 pg/mL, P < 0.05), decreased with LIRA (93 ± 6 to 80 ± 6 pg/mL, P < 0.05), and did not change in CANA/LIRA. LIRA prevented the insulin decline and blocked the glucagon rise observed with CANA but did not inhibit the increase in EGP. Factors other than insulin and glucagon contribute to the stimulation of EGP after CANA-induced glucosuria.


Subject(s)
Canagliflozin/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Down-Regulation/drug effects , Gluconeogenesis/drug effects , Hypoglycemic Agents/therapeutic use , Incretins/therapeutic use , Liraglutide/therapeutic use , Adult , Canagliflozin/adverse effects , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Drug Therapy, Combination/adverse effects , Female , Glucagon/agonists , Glucagon/antagonists & inhibitors , Glucagon/blood , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Humans , Hyperglycemia/prevention & control , Hypoglycemia/chemically induced , Hypoglycemia/prevention & control , Hypoglycemic Agents/adverse effects , Incretins/adverse effects , Insulin/agonists , Insulin/blood , Insulin/chemistry , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Liraglutide/adverse effects , Male , Membrane Transport Modulators/adverse effects , Membrane Transport Modulators/therapeutic use , Middle Aged , Reproducibility of Results , Sodium-Glucose Transporter 2/metabolism , Sodium-Glucose Transporter 2 Inhibitors
8.
Int J Biol Macromol ; 106: 379-386, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28811203

ABSTRACT

Insulin resistance is a key feature of type 2 diabetes mellitus (T2DM) and is characterized by defects in insulin signaling. Protein tyrosine phosphatase 1B (PTP1B) is a major negative regulator of insulin signaling cascade and has attracted intensive investigation in recent T2DM therapy study. BPN, a marine-derived bromophenol compound, was isolated from the red alga Rhodomela confervoides. This study investigated the effects of BPN on the insulin signaling pathway in insulin-resistant C2C12 myotubes by inhibiting PTP1B. Molecular docking study and analysis of small- molecule interaction with PTP1B all showed BPN inhibited PTP1B activity via binding to the catalytic site through hydrogen bonds. We then found that BPN permeated into C2C12 myotubes, on the one hand, activated insulin signaling in an insulin-independent manner in C2C12 cells; on the other hand, ameliorated palmitate-induced insulin resistance through augmenting insulin sensitivity. Moreover, our studies also showed that PTP1B inhibition by BPN increased glucose uptake in normal and insulin-resistant C2C12 myotubes through glucose transporter 4 (GLUT4) translocation. Taken together, BPN activates insulin signaling and alleviates insulin resistance and represents a potential candidate for further development as an antidiabetic agent.


Subject(s)
Hypoglycemic Agents/pharmacology , Insulin/metabolism , Myoblasts/drug effects , Phenols/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Rhodophyta/chemistry , Animals , Catalytic Domain , Cell Line, Transformed , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Hydrogen Bonding , Hypoglycemic Agents/chemistry , Hypoglycemic Agents/isolation & purification , Insulin/agonists , Insulin Resistance , Mice , Molecular Docking Simulation , Myoblasts/metabolism , Myoblasts/pathology , Palmitic Acid/antagonists & inhibitors , Palmitic Acid/pharmacology , Phenols/chemistry , Phenols/isolation & purification , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Tyrosine Phosphatase, Non-Receptor Type 1/chemistry , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction/drug effects
9.
Diabetes Obes Metab ; 20(1): 34-41, 2018 01.
Article in English | MEDLINE | ID: mdl-28544214

ABSTRACT

The prevalence of type 2 diabetes is increasing, which is alarming because of its serious complications. Anti-diabetic treatment aims to control glucose homeostasis as tightly as possible in order to reduce these complications. Dipeptidyl peptidase-4 (DPP-4) inhibitors are a recent addition to the anti-diabetic treatment modalities, and have become widely accepted because of their good efficacy, their benign side-effect profile and their low hypoglycaemia risk. The actions of DPP-4 inhibitors are not direct, but rather are mediated indirectly through preservation of the substrates they protect from degradation. The two incretin hormones, glucagon-like peptide-1 and glucose-dependent insulinotropic polypeptide, are known substrates, but other incretin-independent mechanisms may also be involved. It seems likely therefore that the mechanisms of action of DPP-4 inhibitors are more complex than originally thought, and may involve several substrates and encompass local paracrine, systemic endocrine and neural pathways, which are discussed here.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Incretins/therapeutic use , Models, Biological , Animals , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Gastric Inhibitory Polypeptide/agonists , Gastric Inhibitory Polypeptide/metabolism , Gastrointestinal Motility/drug effects , Glucagon-Like Peptide 1/agonists , Glucagon-Like Peptide 1/metabolism , Humans , Incretins/adverse effects , Incretins/pharmacology , Insulin/agonists , Insulin/metabolism , Insulin Secretion , Pancreas/drug effects , Pancreas/innervation , Pancreas/metabolism , Proteolysis/drug effects , Signal Transduction/drug effects , Synaptic Transmission/drug effects
10.
PLoS One ; 12(8): e0183988, 2017.
Article in English | MEDLINE | ID: mdl-28859155

ABSTRACT

Diabetes is caused by the lack of release or action of insulin. Some foods and supplements can compensate for this deficiency; thus, they can aid in the prevention or treatment of diabetes. The aim of this study was to investigate the effects of Cyclocarya paliurus extract (CPE) on insulin signaling and its capacity to correct hyperglycemia in the absence of insulin. To investigate the hypoglycemic effects of CPE, C2C12 cells were exposed to CPE (50 and 100 µg/mL). CPE promoted 2-(N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)Amino)-2-Deoxyglucose (2NBDG) uptake into the cells via translocation of glucose transporter 4 (Glut4) to the plasma membrane. In addition, CPE enhanced tyrosine phosphorylation of insulin receptor substrate and activated phosphatidylinositol 3-kinase and protein kinase B (Akt) via sirtuin1 in C2C12 cells. Moreover, we found that oral administration of CPE (1 g/kg) to streptozotocin-induced hyperglycemic mice produced a progressive decrease in plasma glucose levels at 1 h after single dosing. At that point, CPE significantly increased the expression of skeletal muscle membrane Glut4 and enhanced the phosphorylation of Akt. These results suggest that CPE exerts antidiabetic effects similar to those of insulin, and may be an oral therapeutic alternative for the management of diabetes.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Drugs, Chinese Herbal/pharmacology , Fagaceae/chemistry , Hypoglycemic Agents/pharmacology , Insulin/agonists , Signal Transduction/drug effects , Sirtuin 1/genetics , 4-Chloro-7-nitrobenzofurazan/analogs & derivatives , 4-Chloro-7-nitrobenzofurazan/metabolism , Animals , Biological Transport/drug effects , Cell Line , Deoxyglucose/analogs & derivatives , Deoxyglucose/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Drugs, Chinese Herbal/isolation & purification , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/isolation & purification , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Mice, Inbred ICR , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phosphorylation/drug effects , Protein Transport , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Sirtuin 1/metabolism
11.
Diabetes Obes Metab ; 19 Suppl 1: 22-29, 2017 09.
Article in English | MEDLINE | ID: mdl-28880474

ABSTRACT

Insulin secretagogues including sulfonylureas, glinides and incretin-related drugs such as dipeptidyl peptidase 4 (DPP-4) inhibitors and glucagon-like peptide-1 receptor agonists are widely used for treatment of type 2 diabetes. In addition, glucokinase activators and G-protein-coupled receptor 40 (GPR40) agonists also have been developed, although the drugs are not clinically usable. These different drugs exert their effects on insulin secretion by different mechanisms. Recent advances in ß-cell signalling studies have not only deepened our understanding of insulin secretion but also revealed novel mechanisms of insulin secretagogues. Clarification of the signalling mechanisms of the insulin secretagogues will contribute to improved drug therapy for diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Drugs, Investigational/therapeutic use , Hypoglycemic Agents/therapeutic use , Insulin-Secreting Cells/drug effects , Insulin/agonists , Models, Biological , Signal Transduction/drug effects , Animals , Diabetes Mellitus, Type 2/metabolism , Drugs, Investigational/adverse effects , Drugs, Investigational/pharmacology , Humans , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacology , Incretins/adverse effects , Incretins/pharmacology , Incretins/therapeutic use , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism
12.
Hum Reprod ; 32(7): 1465-1473, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28482050

ABSTRACT

STUDY QUESTION: Are bisphenol A (BPA) and BPA analogs (BPA-A) safe for male human reproductive function? SUMMARY ANSWER: The endocrine function of human testes explants [assessed by measuring testosterone and insulin-like factor 3 (INSL3)] was impacted by exposure of the human adult testis explants to BPA/BPA-A. WHAT IS KNOWN ALREADY: The few epidemiologic studies performed suggest that bisphenols have potential endocrine disruptive properties, but they did not identify clear and direct patterns of endocrine disruption. STUDY DESIGN, SIZE, DURATION: Adult human testis explants in culture were exposed to BPA and the analogs bisphenol F (BPF), bisphenol S (BPS), bisphenol E (BPE), bisphenol B (BPB) and bisphenol A diglycidyl ether (BADGE) at 10-9-10-5 M for 24 or 48 h. PARTICIPANTS/MATERIALS, SETTING, METHODS: Human adult testes were obtained from prostate cancer patients who had no hormone therapy, or from multiorgan donors. After ex vivo exposure to the investigated bisphenols, the measured outcomes were related to histopathology (gross morphology and germ cell viability determined by anti-caspase three immunohistochemistry), and the levels of testosterone, INSL3 and inhibin B were measured using immunoassays. The levels of mRNA encoding key enzymes of bisphenol biotransformation were investigated by quantitative PCR: UGT2B15 UDP (glucuronosyltransferase two family, polypeptide B15), GUSB (glucuronidase beta), SULT1A1 and 3 (sulfotransferase family 1 A member 1 and 3) and STS (steroid sulfatase). MAIN RESULTS AND THE ROLE OF CHANCE: A significant dose-dependent inhibition was found between testosterone levels measured in the culture medium and concentrations of BPA (P = 0.00778 at 24 h and P = 0.0291 at 48 h), BPE (P = 0.039) and BPF (P = 0.00663). The observed BPA and BPA-A-induced inhibition of testosterone production varied according to duration of exposure and BPA/BPA-A concentrations. BPA (10-9 M; P < 0.05), BPB (10-9 M; P < 0.05), BPS (10-9 and 10-8 M; P < 0.05) and BADGE (10-5 M; P < 0.05) increased Leydig cell INSL3 production. By contrast, BPE dose dependently inhibited INSL3 (P = 0.0372). Conversely, Sertoli cell function (inhibin B) and germ cell viability were not significantly affected by either bisphenols. LARGE SCALE DATA: N/A. LIMITATIONS, REASONS FOR CAUTION: Environmental compounds cannot be deliberately administered to men, justifying the use of an ex vivo approach. A relatively low number of testes samples were available for analysis (n = 3, except for testosterone secretion with n = 5). The active concentrations of BPA and BPA-A used in the study were higher than those found in human biological fluids. WIDER IMPLICATIONS OF THE FINDINGS: Under our experimental conditions, direct exposure to BPA or BPA-A can result in endocrine disturbance in the adult human testis. STUDY FUNDING/COMPETING INTEREST(S): This study was funded by Inserm (Institut National de la Santé et de la Recherche Médicale), EHESP-School of Public Health, University of Rennes1, by grants from the Agence Nationale de la Recherche (ANR; grant#ANR-13-CESA-0012-03 NEWPLAST) and Agence Nationale de Sécurité Sanitaire de l'Alimentation, de l'Environnement et du Travail (ANSES; grant#EST-2010/2/046 (BPATESTIS)). All authors declare they have no current or potential competing financial interests.


Subject(s)
Benzhydryl Compounds/toxicity , Endocrine Disruptors/toxicity , Gene Expression Regulation, Enzymologic/drug effects , Insulin/agonists , Nonsteroidal Anti-Androgens/toxicity , Phenols/toxicity , Proteins/agonists , Testis/drug effects , Testosterone/antagonists & inhibitors , Adult , Apoptosis/drug effects , Arylsulfotransferase/genetics , Arylsulfotransferase/metabolism , Benzhydryl Compounds/chemistry , Endocrine Disruptors/chemistry , Epoxy Compounds/toxicity , Glucuronidase/genetics , Glucuronidase/metabolism , Glucuronosyltransferase/genetics , Glucuronosyltransferase/metabolism , Humans , Insulin/metabolism , Leydig Cells/cytology , Leydig Cells/drug effects , Leydig Cells/metabolism , Male , Nonsteroidal Anti-Androgens/chemistry , Phenols/chemistry , Proteins/antagonists & inhibitors , Proteins/metabolism , Reproducibility of Results , Sertoli Cells/cytology , Sertoli Cells/drug effects , Sertoli Cells/metabolism , Steryl-Sulfatase/genetics , Steryl-Sulfatase/metabolism , Sulfones/toxicity , Testis/cytology , Testis/metabolism , Testosterone/metabolism , Tissue Culture Techniques
13.
J Biol Regul Homeost Agents ; 31(1): 183-188, 2017.
Article in English | MEDLINE | ID: mdl-28337890

ABSTRACT

Protein hydrolysates (PH) are rich sources of proteins that supply the need of exercising muscles. PHs are enriched in di- and tripeptides and are better than free amino acids or intact proteins when muscle anabolic effect is considered. Digestion, absorption and muscle uptake of amino acids are faster and more efficient when PH is ingested in comparison to the respective intact protein. PHs not only enhance endurance in high intensity exercise regimen, but also help in faster post-exercise recovery of muscle by promoting glycogen synthesis, although the latter effect requires more convincing evidence. PHs have been shown to exhibit insulinotrophic effect as it enhances the secretion of insulin and the hormone, in turn, exerts muscle anabolic effect.


Subject(s)
Dietary Proteins/administration & dosage , Physical Endurance/drug effects , Protein Hydrolysates/administration & dosage , Sports Nutritional Sciences , Amino Acids/metabolism , Biological Transport , Dietary Proteins/metabolism , Exercise , Glycogen/biosynthesis , Humans , Insulin/agonists , Insulin/metabolism , Insulin Secretion , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Physical Endurance/physiology , Protein Hydrolysates/metabolism , Sports/physiology
14.
Diabetes Obes Metab ; 19(8): 1088-1096, 2017 08.
Article in English | MEDLINE | ID: mdl-28211608

ABSTRACT

AIMS: 55P0251 is a novel compound with blood glucose lowering activity in mice, which has been developed from a molecular backbone structure found in herbal remedies. We here report its basic pharmacological attributes and initial progress in unmasking the mode of action. MATERIALS AND METHODS: Pharmacokinetic properties of 55P0251 were portrayed in several species. First efforts to elucidate the glucose lowering mechanism in rodents included numerous experimental protocols dealing with glucose tolerance, insulin secretion from isolated pancreatic islets and comparison to established drugs. RESULTS: A single oral dose of 55P0251 improved glucose tolerance in mice with an ED50 between 1.5 and 2 mg/kg (reductions in areas under the curve, 1 mg/kg, -18%; 5 mg/kg, -30%; 27 mg/kg, -47%). Pharmacokinetic studies revealed attractive attributes, including a plasma half-life of approximately 3 hours and a bioavailability of approximately 58% in rats. 55P0251 amplified glucose stimulated insulin release from isolated mouse islets and improved glucose tolerance via increased insulin secretion in rats (increase in area under the insulin curve, +184%). Unlike sulfonylureas and glinides, 55P0251 hardly stimulated insulin release under basal conditions and did not induce hypoglycaemia in vivo, but it amplified the secretory response to glucose and other insulinotropic stimuli (KCl, glucagon-like peptide-1). Comparison to established anti-diabetic agents and examination of interaction with molecular targets (KATP channel, dipeptidyl peptidase-4, glucagon-like peptide-1 receptor) excluded molecular mechanisms addressed by presently marketed drugs. CONCLUSIONS: 55P0251 is a novel compound that potently counteracts hyperglycaemia in rodents via amplification of glucose-stimulated insulin release.


Subject(s)
Alkaloids/therapeutic use , Drugs, Investigational/therapeutic use , Glucose Intolerance/drug therapy , Hyperglycemia/prevention & control , Incretins/therapeutic use , Administration, Oral , Alkaloids/administration & dosage , Alkaloids/pharmacokinetics , Alkaloids/pharmacology , Animals , Biological Availability , Blood Glucose/analysis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Drugs, Investigational/administration & dosage , Drugs, Investigational/pharmacokinetics , Drugs, Investigational/pharmacology , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose Intolerance/blood , Glucose Intolerance/metabolism , Half-Life , Hypoglycemic Agents/pharmacology , Incretins/administration & dosage , Incretins/pharmacokinetics , Incretins/pharmacology , Insulin/agonists , Insulin/blood , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , KATP Channels/antagonists & inhibitors , KATP Channels/metabolism , Male , Metabolic Clearance Rate , Mice, Inbred C57BL , Rats, Sprague-Dawley
15.
J Physiol Biochem ; 72(4): 699-710, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27474043

ABSTRACT

Omega-3 fatty acids have multiple effects in peripheral tissues and pancreatic beta cell function. Dietary depletion of omega-3 fatty acids is associated with pancreatic islet dysfunction and insulin resistance in rats. Herein, the effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on pancreatic beta cell redox state and function were investigated. INS-1E insulin-secreting cells were incubated with EPA and DHA in combination with palmitic acid, and productions of reactive oxygen species (ROS), nitric oxide (NO) and insulin were measured. The involvement of the NADPH oxidase complex in ROS production and expression of the antioxidant enzymes was also investigated. After incubation for 1 or 48 h, productions of superoxide (by hydroethidine method), nitric oxide (by 4,5-diaminofluorescein diacetate-DAF-2DA assay), insulin (by radioimmunoassay), and expressions (by western blot analysis) of glutathione peroxidase (GPx-1) and gp91PHOX were measured. EPA and DHA reduced superoxide production after 1-h incubation. After 48 h, palmitic acid reduced superoxide production that was normalized by EPA treatment. Palmitic acid increased NO production that was reverted by EPA and DHA. Palmitic acid increased insulin secretion after 48 h, whereas both omega-3 fatty acids increased intracellular insulin content. EPA and DHA enhanced GPx-1 expression as well as gp91PHOX glycosylated form. In conclusion, EPA and DHA increased intracellular insulin content and antioxidant enzymatic defense capacity and decreased pro-oxidant generating activities that are associated with maintenance of pancreatic beta cell redox state in response to palmitic acid.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Insulin-Secreting Cells/drug effects , Insulin/biosynthesis , Nitric Oxide/metabolism , Superoxides/metabolism , Animals , Cell Line , Gene Expression Regulation , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Insulin/agonists , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Nitric Oxide/antagonists & inhibitors , Palmitic Acid/antagonists & inhibitors , Palmitic Acid/pharmacology , Rats , Signal Transduction , Superoxides/antagonists & inhibitors , Glutathione Peroxidase GPX1
16.
Appl Biochem Biotechnol ; 180(5): 841-851, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27240663

ABSTRACT

Diabetes has been cited as the most challenging health problem in the twenty-first century. Accordingly, it is urgent to develop a new type of efficient and low-toxic antidiabetic medication. Since vanadium compounds have insulin-mimetic and potential hypoglycemic activities for type 1 and type 2 diabetes, a new trend has been developed using vanadium and organic ligands to form a new compound in order to increase the intestinal absorption and reduce the toxicity of vanadium compound. In the current investigation, a new organic vanadium compounds, vanadyl rosiglitazone, was synthesized and determined by infrared spectra. Vanadyl rosiglitazone and three other organic vanadium compounds were administered to the diabetic mice through oral administration for 5 weeks. The results of mouse model test indicated that vanadyl rosiglitazone could regulate the blood glucose level and relieve the symptoms of polydipsia, polyphagia, polyuria, and weight loss without side effects and was more effective than the other three organic vanadium compounds including vanadyl trehalose, vanadyl metformin, and vanadyl quercetin. The study indicated that vanadyl rosiglitazone presents insulin-mimetic activities, and it will be a good potential candidate for the development of a new type of oral drug for type 2 diabetes.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/drug therapy , Homeostasis , Insulin/agonists , Thiazolidinediones/therapeutic use , Vanadium/therapeutic use , Animals , Body Weight/drug effects , Disease Models, Animal , Drinking Behavior/drug effects , Feeding Behavior/drug effects , Homeostasis/drug effects , Male , Metformin/pharmacology , Metformin/therapeutic use , Mice , Quercetin/pharmacology , Quercetin/therapeutic use , Rosiglitazone , Spectrophotometry, Infrared , Thiazolidinediones/pharmacology , Trehalose/pharmacology , Trehalose/therapeutic use , Vanadium/pharmacology
17.
PLoS One ; 11(4): e0154452, 2016.
Article in English | MEDLINE | ID: mdl-27104960

ABSTRACT

GPR142 is an islet-enriched G protein-coupled receptor that has been investigated as a novel therapeutic target for the treatment of type 2 diabetes by virtue of its insulin secretagogue activity. However, the signaling pathways downstream of GPR142 and whether its stimulation of insulin release is glucose-dependent remain poorly characterized. In this study, we show that both native and synthetic GPR142 agonists can activate Gq as well as Gi signaling when GPR142 is recombinantly expressed in HEK293 cells. However, in primary pancreatic islets, a native cellular system, the insulin secretagogue activity of GPR142 agonists only requires Gq activation. In addition, our results show that stimulation of insulin secretion by GPR142 in pancreatic islets is strictly glucose-dependent.


Subject(s)
Aminopyridines/pharmacology , Glucose/pharmacology , Insulin/agonists , Protein Subunits/agonists , Pyrazoles/pharmacology , Receptors, G-Protein-Coupled/agonists , Tryptophan/pharmacology , Animals , Colforsin/pharmacology , Cyclic AMP/metabolism , Gene Expression Regulation , Glucose/metabolism , HEK293 Cells , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation , Primary Cell Culture , Protein Subunits/genetics , Protein Subunits/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
18.
Diabetes ; 65(5): 1398-409, 2016 05.
Article in English | MEDLINE | ID: mdl-26868296

ABSTRACT

Discovery of common pathways that mediate both pancreatic ß-cell function and end-organ function offers the opportunity to develop therapies that modulate glucose homeostasis and separately slow the development of diabetes complications. Here, we investigated the in vitro and in vivo effects of pharmacological agonism of the prostaglandin I2 (IP) receptor in pancreatic ß-cells and in glomerular podocytes. The IP receptor agonist MRE-269 increased intracellular 3',5'-cyclic adenosine monophosphate (cAMP), augmented glucose-stimulated insulin secretion (GSIS), and increased viability in MIN6 ß-cells. Its prodrug form, selexipag, augmented GSIS and preserved islet ß-cell mass in diabetic mice. Determining that this preservation of ß-cell function is mediated through cAMP/protein kinase A (PKA)/nephrin-dependent pathways, we found that PKA inhibition, nephrin knockdown, or targeted mutation of phosphorylated nephrin tyrosine residues 1176 and 1193 abrogated the actions of MRE-269 in MIN6 cells. Because nephrin is important to glomerular permselectivity, we next set out to determine whether IP receptor agonism similarly affects nephrin phosphorylation in podocytes. Expression of the IP receptor in podocytes was confirmed in cultured cells by immunoblotting and quantitative real-time PCR and in mouse kidneys by immunogold electron microscopy, and its agonism 1) increased cAMP, 2) activated PKA, 3) phosphorylated nephrin, and 4) attenuated albumin transcytosis. Finally, treatment of diabetic endothelial nitric oxide synthase knockout mice with selexipag augmented renal nephrin phosphorylation and attenuated albuminuria development independently of glucose change. Collectively, these observations describe a pharmacological strategy that posttranslationally modifies nephrin and the effects of this strategy in the pancreas and in the kidney.


Subject(s)
Diabetic Nephropathies/prevention & control , Insulin-Secreting Cells/drug effects , Membrane Proteins/metabolism , Podocytes/drug effects , Receptors, Epoprostenol/agonists , Acetamides/therapeutic use , Acetates/pharmacology , Animals , Cell Line , Cell Survival/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Diabetic Nephropathies/physiopathology , Humans , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/therapeutic use , Insulin/agonists , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phosphorylation/drug effects , Podocytes/metabolism , Podocytes/pathology , Podocytes/ultrastructure , Prodrugs/therapeutic use , Protein Processing, Post-Translational/drug effects , Pyrazines/pharmacology , Pyrazines/therapeutic use , RNA Interference , Receptors, Epoprostenol/genetics , Receptors, Epoprostenol/metabolism , Renal Insufficiency/complications , Renal Insufficiency/metabolism , Renal Insufficiency/pathology , Renal Insufficiency/prevention & control
19.
J Med Chem ; 59(5): 2118-25, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26824523

ABSTRACT

Insulin-like peptide 5 (INSL5) has recently been discovered as only the second orexigenic gut hormone after ghrelin. As we have previously reported, INSL5 is extremely difficult to assemble and oxidize into its two-chain three-disulfide structure. The focus of this study was to generate structure-activity relationships (SARs) of INSL5 and use it to develop a potent and simpler INSL5 mimetic with RXFP4 agonist activity. A series of human and mouse INSL5 (hINSL5/mINSL5) analogues were designed and chemically synthesized, resulting in a chimeric INSL5 analogue exhibiting more than 10-fold higher potency (0.35 nM) at human RXFP4 compared with native hINSL5 (4.57 nM). The SAR study also identified a key residue (K(A15)) in the A-chain of mINSL5 that contributes to improved RXFP4 affinity and potency of mINSL5 compared with hINSL5. This knowledge ultimately led us to engineer a minimized hINSL5 mimetic agonist that retains native hINSL5-like RXFP4 affinity and potency at human RXFP4. This minimized analogue was synthesized in 17.5-fold higher yield and in less time compared with hINSL5.


Subject(s)
Insulin/agonists , Peptides/pharmacology , Protein Engineering , Proteins/agonists , Animals , CHO Cells , Cricetulus , Dose-Response Relationship, Drug , Humans , Mice , Models, Molecular , Molecular Structure , Peptides/chemical synthesis , Peptides/chemistry , Structure-Activity Relationship
20.
Physiol Res ; 64(Suppl 2): S237-46, 2015.
Article in English | MEDLINE | ID: mdl-26680485

ABSTRACT

Determination of response of cortisol and its metabolites to different stimuli may be important for adrenal gland disorders. To date, only one metabolite, cortisone, has been followed in stimulation tests of the adrenal gland. We aimed to describe a response of cortisol metabolites to the standard short Synacthen test (HDST), insulin tolerance test (ITT), low dose Synacthen test (LDST) and medium dose Synacthen test (MDST). Sixty healthy subjects were investigated: 30 men and 30 women. Plasma for measurements of cortisol and its metabolites was obtained before and 30th and 60th min after Synacthen and insulin administration. The cut-off 500 nmol/l of cortisol was reached after stimulation in all of tests, the maximal stimulation level was reached in 60th min in all of the tests except for LDST. The response of cortisol and its metabolites at 30th and 60th min strongly correlated in all of the tests except for LDST. Cortisol and its metabolites increased after stimulation; in contrast, cortisone and its metabolites decreased. We showed that the response of the cortisol metabolites during the Synacthen tests and ITT well correlated, and the MDST showed similar response compared to HDST. The decrease in cortisone metabolites may correspond to the regeneration of cortisol from cortisone in response to stimulation test.


Subject(s)
Cortisone/blood , Cosyntropin/pharmacology , Hydrocortisone/blood , Insulin Resistance/physiology , Insulin/blood , Adult , Cortisone/agonists , Female , Humans , Hydrocortisone/agonists , Insulin/agonists , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...