Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
Add more filters

Publication year range
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
J Biol Chem ; 290(34): 21131-21140, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26157145

ABSTRACT

FFAR1/GPR40 is a seven-transmembrane domain receptor (7TMR) expressed in pancreatic ß cells and activated by FFAs. Pharmacological activation of GPR40 is a strategy under consideration to increase insulin secretion in type 2 diabetes. GPR40 is known to signal predominantly via the heterotrimeric G proteins Gq/11. However, 7TMRs can also activate functionally distinct G protein-independent signaling via ß-arrestins. Further, G protein- and ß-arrestin-based signaling can be differentially modulated by different ligands, thus eliciting ligand-specific responses ("biased agonism"). Whether GPR40 engages ß-arrestin-dependent mechanisms and is subject to biased agonism is unknown. Using bioluminescence resonance energy transfer-based biosensors for real-time monitoring of cell signaling in living cells, we detected a ligand-induced GPR40-ß-arrestin interaction, with the synthetic GPR40 agonist TAK-875 being more effective than palmitate or oleate in recruiting ß-arrestins 1 and 2. Conversely, TAK-875 acted as a partial agonist of Gq/11-dependent GPR40 signaling relative to both FFAs. Pharmacological blockade of Gq activity decreased FFA-induced insulin secretion. In contrast, knockdown or genetic ablation of ß-arrestin 2 in an insulin-secreting cell line and mouse pancreatic islets, respectively, uniquely attenuated the insulinotropic activity of TAK-875, thus providing functional validation of the biosensor data. Collectively, these data reveal that in addition to coupling to Gq/11, GPR40 is functionally linked to a ß-arrestin 2-mediated insulinotropic signaling axis. These observations expose previously unrecognized complexity for GPR40 signal transduction and may guide the development of biased agonists showing improved clinical profile in type 2 diabetes.


Subject(s)
Arrestins/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Animals , Arrestins/antagonists & inhibitors , Arrestins/metabolism , Benzofurans/pharmacology , Biosensing Techniques , Cell Line, Tumor , Electron Spin Resonance Spectroscopy , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Gene Expression Regulation , HEK293 Cells , Humans , Insulin/agonists , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Kinetics , Mice , Oleic Acid/pharmacology , Palmitic Acid/pharmacology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Receptors, G-Protein-Coupled/metabolism , Sulfones/pharmacology , Tissue Culture Techniques , beta-Arrestin 2 , beta-Arrestins
11.
J Biol Chem ; 290(34): 20934-20946, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26139601

ABSTRACT

The tumor suppressor liver kinase B1 (LKB1) is an important regulator of pancreatic ß cell biology. LKB1-dependent phosphorylation of distinct AMPK (adenosine monophosphate-activated protein kinase) family members determines proper ß cell polarity and restricts ß cell size, total ß cell mass, and glucose-stimulated insulin secretion (GSIS). However, the full spectrum of LKB1 effects and the mechanisms involved in the secretory phenotype remain incompletely understood. We report here that in the absence of LKB1 in ß cells, GSIS is dramatically and persistently improved. The enhancement is seen both in vivo and in vitro and cannot be explained by altered cell polarity, increased ß cell number, or increased insulin content. Increased secretion does require membrane depolarization and calcium influx but appears to rely mostly on a distal step in the secretion pathway. Surprisingly, enhanced GSIS is seen despite profound defects in mitochondrial structure and function in LKB1-deficient ß cells, expected to greatly diminish insulin secretion via the classic triggering pathway. Thus LKB1 is essential for mitochondrial homeostasis in ß cells and in parallel is a powerful negative regulator of insulin secretion. This study shows that ß cells can be manipulated to enhance GSIS to supra-normal levels even in the face of defective mitochondria and without deterioration over months.


Subject(s)
Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Mitochondria/metabolism , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Gene Expression Regulation , Glucose/pharmacology , Glutamic Acid/metabolism , Humans , Insulin/agonists , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Mice , Mice, Transgenic , Mitochondria/drug effects , Mitochondria/pathology , Phosphorylation , Protein Serine-Threonine Kinases/deficiency , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tamoxifen/toxicity , Tissue Culture Techniques
12.
Dev Biol ; 394(2): 314-26, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128585

ABSTRACT

Caenorhabditis elegans larvae reversibly arrest development in the first larval stage in response to starvation (L1 arrest or L1 diapause). Insulin-like signaling is a critical regulator of L1 arrest. However, the C. elegans genome encodes 40 insulin-like peptides, and it is unknown which peptides participate in nutritional control of L1 development. Work in other contexts has revealed that insulin-like genes can promote development ("agonists") or developmental arrest ("antagonists"), suggesting that such agonists promote L1 development in response to feeding. We measured mRNA expression dynamics with high temporal resolution for all 40 insulin-like genes during entry into and recovery from L1 arrest. Nutrient availability influences expression of the majority of insulin-like genes, with variable dynamics suggesting complex regulation. We identified thirteen candidate agonists and eight candidate antagonists based on expression in response to nutrient availability. We selected ten candidate agonists (daf-28, ins-3, ins-4, ins-5, ins-6, ins-7, ins-9, ins-26, ins-33 and ins-35) for further characterization in L1 stage larvae. We used destabilized reporter genes to determine spatial expression patterns. Expression of candidate agonists is largely overlapping in L1 stage larvae, suggesting a role of the intestine, chemosensory neurons ASI and ASJ, and the interneuron PVT in control of L1 development. Transcriptional regulation of candidate agonists is most significant in the intestine, as if internal nutrient status is a more important influence on transcription than sensory perception. Phenotypic analysis of single and compound deletion mutants did not reveal effects on L1 developmental dynamics, though simultaneous disruption of ins-4 and daf-28 increases survival of L1 arrest. Furthermore, overexpression of ins-4, ins-6 or daf-28 alone decreases survival and promotes cell division during starvation. These results suggest extensive functional overlap among insulin-like genes in nutritional control of L1 development while highlighting the role of ins-4, daf-28 and to a lesser extent ins-6.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental/physiology , Peptide Hormones/metabolism , RNA, Messenger/metabolism , Receptor, Insulin/metabolism , Analysis of Variance , Animals , Caenorhabditis elegans Proteins/genetics , Gene Expression Profiling , Insulin/agonists , Insulin/metabolism , Insulins , Intestinal Mucosa/metabolism , Larva/growth & development , Larva/metabolism , Logistic Models , Peptide Hormones/genetics , Survival Analysis
13.
Am J Physiol Endocrinol Metab ; 308(4): E294-305, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25491723

ABSTRACT

Insulin stimulates glucose uptake in adult skeletal muscle by promoting the translocation of GLUT4 glucose transporters to the transverse tubule (T-tubule) membranes, which have particularly high cholesterol levels. We investigated whether T-tubule cholesterol content affects insulin-induced glucose transport. Feeding mice a high-fat diet (HFD) for 8 wk increased by 30% the T-tubule cholesterol content of triad-enriched vesicular fractions from muscle tissue compared with triads from control mice. Additionally, isolated muscle fibers (flexor digitorum brevis) from HFD-fed mice showed a 40% decrease in insulin-stimulated glucose uptake rates compared with fibers from control mice. In HFD-fed mice, four subcutaneous injections of MßCD, an agent reported to extract membrane cholesterol, improved their defective glucose tolerance test and normalized their high fasting glucose levels. The preincubation of isolated muscle fibers with relatively low concentrations of MßCD increased both basal and insulin-induced glucose uptake in fibers from controls or HFD-fed mice and decreased Akt phosphorylation without altering AMPK-mediated signaling. In fibers from HFD-fed mice, MßCD improved insulin sensitivity even after Akt or CaMK II inhibition and increased membrane GLUT4 content. Indinavir, a GLUT4 antagonist, prevented the stimulatory effects of MßCD on glucose uptake. Addition of MßCD elicited ryanodine receptor-mediated calcium signals in isolated fibers, which were essential for glucose uptake. Our findings suggest that T-tubule cholesterol content exerts a critical regulatory role on insulin-stimulated GLUT4 translocation and glucose transport and that partial cholesterol removal from muscle fibers may represent a useful strategy to counteract insulin resistance.


Subject(s)
Anticholesteremic Agents/therapeutic use , Glucose Transporter Type 4/agonists , Glucose/metabolism , Insulin Resistance , Muscle Fibers, Skeletal/drug effects , Obesity/drug therapy , beta-Cyclodextrins/therapeutic use , Animals , Anticholesteremic Agents/pharmacology , Biological Transport/drug effects , Calcium-Calmodulin-Dependent Protein Kinase Type 2/antagonists & inhibitors , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cells, Cultured , Cholesterol/metabolism , Glucose Transporter Type 4/antagonists & inhibitors , Glucose Transporter Type 4/metabolism , Hypoglycemic Agents/agonists , Hypoglycemic Agents/pharmacology , Insulin/agonists , Insulin/pharmacology , Male , Membrane Transport Modulators/pharmacology , Mice, Inbred C57BL , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Obesity/metabolism , Obesity/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Ryanodine Receptor Calcium Release Channel/drug effects , Ryanodine Receptor Calcium Release Channel/metabolism , beta-Cyclodextrins/pharmacology
14.
Diabetes Metab Res Rev ; 31(6): 610-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25773712

ABSTRACT

BACKGROUND: Lixisenatide is a once-daily, prandial, short-acting glucagon-like peptide-1 receptor agonist. Its main antidiabetic effect is to delay gastric emptying to control postprandial plasma glucose excursions. The dose-response relationship of the integrated insulinotropic and gastrostatic response to lixisenatide in healthy volunteers after a standardized liquid meal was investigated. METHODS: Twenty healthy subjects received acetaminophen 1000 mg with a standardized liquid meal 60 min after a single subcutaneous injection of placebo or lixisenatide 2.5, 5, 10 or 20 µg in randomized order separated by a 2- to 7-day washout. Acetaminophen pharmacokinetics served as a surrogate to assess rate of gastric emptying. Postprandial plasma glucose, insulin, C-peptide and glucagon were assessed for 5 h after the meal test, and lixisenatide pharmacokinetics were determined for 6 h. RESULTS: After lixisenatide administration and prior to the standardized meal, insulin and C-peptide transiently increased, while fasting plasma glucose decreased in a dose-dependent manner. After the meal, postprandial plasma glucose, insulin and C-peptide were dose proportionally reduced with lixisenatide versus placebo for up to 6 h. Compared with placebo, glucagon levels were transiently lower after any lixisenatide dose, with more sustained reductions after the meal and no apparent dose-related trends. Acetaminophen absorption was significantly reduced and delayed compared with placebo for lixisenatide doses ≥5 µg and demonstrated dose-dependent slowing of gastric emptying. Lixisenatide displayed near dose-proportional exposure, with gastrointestinal events increasing with dose. CONCLUSIONS: Lixisenatide reduced fasting plasma glucose via stimulation of glucose-dependent insulin release and controlled postprandial plasma glucose by delaying gastric emptying, demonstrating it to be a valuable option for overall glycaemic control.


Subject(s)
Gastric Emptying/drug effects , Gastrointestinal Agents/pharmacokinetics , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacokinetics , Insulin/agonists , Peptides/pharmacokinetics , Acetaminophen/blood , Acetaminophen/pharmacokinetics , Adult , Analgesics, Non-Narcotic/blood , Analgesics, Non-Narcotic/pharmacokinetics , Blood Glucose/analysis , C-Peptide/blood , Cross-Over Studies , Dose-Response Relationship, Drug , Female , Gastrointestinal Agents/blood , Gastrointestinal Agents/pharmacology , Glucagon/blood , Glucagon/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/blood , Injections, Subcutaneous , Insulin/blood , Insulin/metabolism , Insulin Secretion , Intestinal Absorption/drug effects , Male , Peptides/administration & dosage , Peptides/blood , Postprandial Period , Young Adult
15.
J Nutr ; 145(10): 2325-32, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26338890

ABSTRACT

BACKGROUND: Dietary cocoa is an important source of flavonoids and is associated with favorable cardiovascular disease effects, such as improvements in vascular function and lipid profiles, in nondiabetic adults. Type 2 diabetes (T2D) is associated with adverse effects on postprandial serum glucose, lipids, inflammation, and vascular function. OBJECTIVE: We examined the hypothesis that cocoa reduces metabolic stress in obese T2D adults after a high-fat fast-food-style meal. METHODS: Adults with T2D [n = 18; age (mean ± SE): 56 ± 3 y; BMI (in kg/m(2)): 35.3 ± 2.0; 14 women; 4 men] were randomly assigned to receive cocoa beverage (960 mg total polyphenols; 480 mg flavanols) or flavanol-free placebo (110 mg total polyphenols; <0.1 mg flavanols) with a high-fat fast-food-style breakfast [766 kcal, 50 g fat (59% energy)] in a crossover trial. After an overnight fast (10-12 h), participants consumed the breakfast with cocoa or placebo, and blood sample collection [glucose, insulin, lipids, and high-sensitivity C-reactive protein (hsCRP)] and vascular measurements were conducted at 0.5, 1, 2, 4, and 6 h postprandially on each study day. Insulin resistance was evaluated by homeostasis model assessment. RESULTS: Over the 6-h study, and specifically at 1 and 4 h, cocoa increased HDL cholesterol vs. placebo (overall Δ: 1.5 ± 0.8 mg/dL; P ≤ 0.01) but had no effect on total and LDL cholesterol, triglycerides, glucose, and hsCRP. Cocoa increased serum insulin concentrations overall (Δ: 5.2 ± 3.2 mU/L; P < 0.05) and specifically at 4 h but had no overall effects on insulin resistance (except at 4 h, P < 0.05), systolic or diastolic blood pressure, or small artery elasticity. However, large artery elasticity was overall lower after cocoa vs. placebo (Δ: -1.6 ± 0.7 mL/mm Hg; P < 0.05), with the difference significant only at 2 h. CONCLUSION: Acute cocoa supplementation showed no clear overall benefit in T2D patients after a high-fat fast-food-style meal challenge. Although HDL cholesterol and insulin remained higher throughout the 6-h postprandial period, an overall decrease in large artery elasticity was found after cocoa consumption. This trial was registered at clinicaltrials.gov as NCT01886989.


Subject(s)
Beverages , Cacao , Cholesterol, HDL/agonists , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/prevention & control , Insulin/agonists , Obesity/complications , Beverages/adverse effects , Beverages/analysis , Body Mass Index , Breakfast , Cacao/adverse effects , Cacao/chemistry , Cholesterol, HDL/analysis , Cross-Over Studies , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetic Angiopathies/complications , Diet, High-Fat/adverse effects , Double-Blind Method , Female , Flavonoids/adverse effects , Flavonoids/analysis , Flavonoids/therapeutic use , Humans , Insulin/blood , Insulin Resistance , Male , Middle Aged , Postprandial Period , Vascular Stiffness
16.
Bioorg Med Chem Lett ; 24(14): 3100-3, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24881566

ABSTRACT

The exploration of a diarylsulfonamide series of free fatty acid receptor 4 (FFA4/GPR120) agonists is described. This work led to the identification of selective FFA4 agonist 8 (GSK137647A) and selective FFA4 antagonist 39. The in vitro profile of compounds 8 and 39 is presented herein.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Sulfonamides/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Insulin/agonists , Mice , Molecular Structure , Structure-Activity Relationship , Sulfonamides/chemical synthesis , Sulfonamides/chemistry
17.
Molecules ; 19(4): 4654-68, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24739928

ABSTRACT

A physio-pathological feature of diabetes mellitus is a significant reduction of ß-pancreatic cells. The growth, differentiation and function maintenance of these cells is directed by transcription factors. Nkx6.1 is a key transcription factor for the differentiation, neogenesis and maintenance of ß-pancreatic cells. We reported that silymarin restores normal morphology and endocrine function of damaged pancreatic tissue after alloxan-induced diabetes mellitus in rats. The aim of this study was to analyze the effect of silymarin on Nkx6.1 transcription factor expression and its consequence in ß cells neogenesis. Sixty male Wistar rats were partially pancreatectomized and divided into twelve groups. Six groups were treated with silymarin (200 mg/Kg p.o) for periods of 3, 7, 14, 21, 42 and 63 days. Additionally, an unpancreatectomized control group was used. Nkx6.1 and insulin gene expression were assessed by RT-PCR assay in total pancreatic RNA. ß-Cell neogenesis was determined by immunoperoxidase assay. Silymarin treated group showed an increase of Nkx6.1 and insulin genic expression. In this group, there was an increment of ß-cell neogenesis in comparison to pancreatectomized untreated group. Silymarin treatment produced a rise in serum insulin and serum glucose normalization. These results suggest that silymarin may improve the reduction of ß pancreatic cells observed in diabetes mellitus.


Subject(s)
Homeodomain Proteins/agonists , Insulin-Secreting Cells/drug effects , Insulin/agonists , Pancreatectomy , Protective Agents/pharmacology , Silymarin/pharmacology , Animals , Blood Glucose/metabolism , Cell Count , Cell Proliferation , Diabetes Mellitus , Disease Models, Animal , Gene Expression , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Immunoenzyme Techniques , Insulin/genetics , Insulin/metabolism , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Male , Rats , Rats, Wistar
18.
Eksp Klin Farmakol ; 77(5): 6-9, 2014.
Article in Russian | MEDLINE | ID: mdl-25033564

ABSTRACT

The dependence of the antidepressant action of 3-oxypyridine and succinic acid derivatives (emoxipine, reamberin, and mexidol) on the insulin potentiating activity of these therapeutic agents has been studied in experiments on rats. Alpha-lipoic acid was used as a reference drug. It was established that single administration of emoxipine, reamberin, mexidol and alpha-lipoic acid in optimal doses, corresponding to the therapeutic range in humans, increased the sensitivity of animals to insulin according to the criterion of insulin coma development. Triple administration of the therapeutic agents studied in the same single dose produced an antidepressant effect according to the criterion of "desperate behavior" in Porsolt forced swimming test. Standardization of obtained data by average difference from the control and further correlation analysis demonstrated that the extent of antidepressant action of emoxipine, reamberin, mexidol and alpha-lipoic acid considerably depends on their insulin potentiating activity (r = 0.762, p = 0.004).


Subject(s)
Antidepressive Agents/pharmacology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Pyridines/pharmacology , Succinates/pharmacology , Animals , Antidepressive Agents/agonists , Female , Hypoglycemic Agents/agonists , Insulin/agonists , Male , Pyridines/agonists , Rats
19.
Pharmacogenet Genomics ; 23(9): 479-86, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23873118

ABSTRACT

OBJECTIVES: Sympathetic activation inhibits insulin secretion through activation of pancreatic α(2)A adrenoreceptors (α(2A)ARs). A common genetic α(2A)AR variant (rs553668) is associated with impaired insulin secretion. α(2A)R agonists would be expected to decrease insulin secretion, but their effects on glucose homeostasis in humans are poorly characterized. We examined the hypotheses that the selective α(2A)R agonist, dexmedetomidine, decreases plasma insulin levels and increases plasma glucose levels in humans and that these effects are modified by genetic α(2A)AR variants. METHODS: Healthy, fasting, White (n=31) and Black (n=33) participants aged between 18 and 45 years received three sequential infusions of placebo (normal saline) at 30-min intervals, followed by three infusions of dexmedetomidine (0.1, 0.15, and 0.15 mcg/kg). Plasma insulin and glucose concentrations were measured at baseline and after the administration of placebo and dexmedetomidine. We genotyped ADRA2A rs553668 and rs2484516, which characterize haplotypes 4 and 4b, respectively. RESULTS: Dexmedetomidine decreased fasting insulin concentrations by 37%, from a median value after placebo administration of 7.9 µU/ml (interquartile range: 6.0-12.6) to 4.9 µU/ml (interquartile range: 3.5-7.9; P<0.001). Plasma glucose concentrations increased from 76±6 to 79±7 mg/dl (P<0.001). The rs2484516 variant allele was associated with higher baseline insulin concentrations before (P=0.001) and after adjustment for potential confounders (P=0.014) and a greater decrease in insulin concentration after dexmedetomidine administration (P=0.016), which was no longer significant after adjustment for baseline concentrations and other confounders (P=0.58). CONCLUSION: Low-dose dexmedetomidine decreased plasma insulin concentration and mildly increased plasma glucose concentration in healthy fasting individuals. The ADRA2A genetic variation may affect baseline insulin concentrations and thus the insulin decrease after dexmedetomidine administration.


Subject(s)
Blood Glucose/metabolism , Dexmedetomidine/pharmacology , Insulin/metabolism , Polymorphism, Single Nucleotide , Receptors, Adrenergic, alpha-2/genetics , Adolescent , Adult , Cohort Studies , Dexmedetomidine/administration & dosage , Dexmedetomidine/agonists , Dexmedetomidine/therapeutic use , Drug Administration Schedule , Female , Genetic Variation , Genotyping Techniques , Haplotypes , Humans , Insulin/agonists , Insulin/blood , Insulin Secretion , Middle Aged , Single-Blind Method , Young Adult
20.
Diabetes Obes Metab ; 15(7): 650-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23388064

ABSTRACT

AIM: The incretin hormones, glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) and cholecystokinin (CCK) are gastrointestinal peptides with important physiological effects. However, rapid enzymatic degradation results in short-lived biological actions. METHODS: This study has examined metabolic actions of exendin-4, GIP[mPEG] and a novel CCK-8 analogue, (pGlu-Gln)-CCK-8 as enzymatically stable forms of GLP-1, GIP and CCK, respectively. RESULTS: All peptides significantly (p < 0.01-p < 0.001) stimulated insulin secretion from BRIN BD11 cells, and acute in vivo experiments confirmed prominent antihyperglycaemic and insulinotropic responses to GLP-1 or GIP receptor activation in normal mice. Twice daily injection of (pGlu-Gln)-CCK-8 alone and in combination with exendin-4 or GIP[mPEG] in high fat-fed mice significantly decreased accumulated food intake (p < 0.05-p < 0.01), body weight gain (p < 0.05-p < 0.01) and improved (p < 0.05) insulin sensitivity in high fat-fed mice. However, there was no evidence for superior effects compared to (pGlu-Gln)-CCK-8 alone. Combined treatment of (pGlu-Gln)-CCK-8 and exendin-4 resulted in significantly (p < 0.05) lowered circulating glucose levels and improved (p < 0.05) intraperitoneal glucose tolerance. These effects were superior to either treatment regime alone but not associated with altered insulin concentrations. A single injection of (pGlu-Gln)-CCK-8, or combined with exendin-4, significantly (p < 0.05) lowered blood glucose levels 24 h post injection in untreated high fat-fed mice. CONCLUSION: This study highlights the potential of (pGlu-Gln)-CCK-8 alone and in combination with incretin hormones for the treatment of type 2 diabetes.


Subject(s)
Anti-Obesity Agents/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Gastric Inhibitory Polypeptide/analogs & derivatives , Hypoglycemic Agents/therapeutic use , Obesity/drug therapy , Peptides/therapeutic use , Sincalide/analogs & derivatives , Venoms/therapeutic use , Animals , Anti-Obesity Agents/pharmacology , Appetite Regulation/drug effects , Cell Line , Diabetes Mellitus, Type 2/complications , Diet, High-Fat/adverse effects , Drug Therapy, Combination , Exenatide , Gastric Inhibitory Polypeptide/pharmacology , Gastric Inhibitory Polypeptide/therapeutic use , Hyperglycemia/prevention & control , Hypoglycemic Agents/pharmacology , Incretins/pharmacology , Incretins/therapeutic use , Insulin/agonists , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred Strains , Obesity/complications , Obesity/etiology , Peptides/pharmacology , Rats , Sincalide/pharmacology , Sincalide/therapeutic use , Time Factors , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL