Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
Add more filters

Complementary Medicines
Country/Region as subject
Publication year range
1.
Science ; 376(6599): 1321-1327, 2022 06 17.
Article in English | MEDLINE | ID: mdl-35709255

ABSTRACT

The emergence of new therapeutic modalities requires complementary tools for their efficient syntheses. Availability of methodologies for site-selective modification of biomolecules remains a long-standing challenge, given the inherent complexity and the presence of repeating residues that bear functional groups with similar reactivity profiles. We describe a bioconjugation strategy for modification of native peptides relying on high site selectivity conveyed by enzymes. We engineered penicillin G acylases to distinguish among free amino moieties of insulin (two at amino termini and an internal lysine) and manipulate cleavable phenylacetamide groups in a programmable manner to form protected insulin derivatives. This enables selective and specific chemical ligation to synthesize homogeneous bioconjugates, improving yield and purity compared to the existing methods, and generally opens avenues in the functionalization of native proteins to access biological probes or drugs.


Subject(s)
Insulin , Penicillin Amidase , Peptides , Protein Engineering , Amino Acid Sequence , Humans , Insulin/analogs & derivatives , Insulin/biosynthesis , Lysine/chemistry , Penicillin Amidase/chemistry , Penicillin Amidase/genetics , Peptides/chemistry , Peptides/genetics , Protein Engineering/methods
2.
Biomed Pharmacother ; 146: 112552, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34923339

ABSTRACT

Blood glucose is inadequately controlled in diabetes mellitus, causing various inflammation-related complications. This study aimed to investigate responses to an oral sucrose/lipid challenge in the context of glucose metabolism after consumption of Mori ramulus (MR) extract. In this study on healthy subjects, the optimal dose and safety of MR were confirmed in a preliminary pilot trial (n = 24), meanwhile, blood glucose, insulin, and inflammatory marker levels were detected via an oral sucrose/lipid tolerance test in the main trial (n = 36). In the main study, the blood glucose response was significantly decreased after 240 min in the MR group. Compared to the placebo group, the treatment group exhibited plasma insulin levels that were significantly increased at 120 min and decreased at 240 min. In conclusion, a single MR extract dose protects against inflammation induced by high-fat/sugar to maintain normal insulin secretion and thus helps to maintain postprandial blood glucose levels via an inflammatory mechanism.


Subject(s)
Blood Glucose/drug effects , Inflammation Mediators/metabolism , Morus , Plant Extracts/pharmacology , Adult , Chemokines/drug effects , Cross-Over Studies , Cytokines/drug effects , Diet, High-Fat , Double-Blind Method , Female , Humans , Insulin/biosynthesis , Male , Postprandial Period , Young Adult
3.
Curr Drug Metab ; 22(13): 1017-1034, 2021.
Article in English | MEDLINE | ID: mdl-34825869

ABSTRACT

The N-3 polyunsaturated fatty acids (PUFAs) have a wide range of health benefits, including antiinflammatory effects, improvements in lipids metabolism and promoting insulin secretion, as well as reduction of cancer risk. Numerous studies support that N-3 PUFAs have the potentials to improve many metabolic diseases, such as diabetes, nonalcoholic fatty liver disease and obesity, which are attributable to N-3 PUFAs mediated enhancement of insulin secretion by pancreatic ß-cells and improvements in insulin sensitivity and metabolic disorders in peripheral insulin-sensitive tissues such as liver, muscles, and adipose tissue. In this review, we summarized the up-to-date clinical and basic studies on the regulatory effects and molecular mechanisms of N-3 PUFAs mediated benefits on pancreatic ß-cells, adipose tissue, liver, and muscles in the context of glucose and/or lipid metabolic disorders. We also discussed the potential factors involved in the inconsistent results from different clinical researches of N-3 PUFAs.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Insulin-Secreting Cells , Insulin , Lipid Metabolism/drug effects , Metabolic Diseases , Animals , Fatty Acids, Omega-3/metabolism , Fatty Acids, Omega-3/pharmacology , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin Resistance/physiology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Metabolic Diseases/classification , Metabolic Diseases/metabolism , Metabolic Diseases/prevention & control
4.
J Immunol Res ; 2020: 9680508, 2020.
Article in English | MEDLINE | ID: mdl-32566690

ABSTRACT

Chlorogenic acid (CA) is a phenolic compound commonly found in human plant-based diets. CA is the main component of many traditional Chinese medicine preparations, and in recent years, it has been found to have hypoglycemic, hypolipidemic, anti-inflammatory, antioxidant, and other pharmacological properties. Specifically, CA relieves the effects of, and prevents, diabetes mellitus (DM). In addition, CA is also beneficial against complications arising from DM, such as diabetic nephropathy (DN), diabetic retinopathy (DR), and diabetic peripheral neuropathy (DPN). Herein, we review the use of CA in the prevention and treatment of DM and its complications, providing a background for further research and medical uses.


Subject(s)
Chlorogenic Acid/administration & dosage , Diabetes Complications/drug therapy , Diabetes Mellitus/drug therapy , Hypoglycemic Agents/administration & dosage , Animals , Antioxidants/administration & dosage , Diabetes Complications/prevention & control , Diabetes Mellitus/prevention & control , Gene Expression Regulation, Enzymologic/drug effects , Glucose/metabolism , Humans , Insulin/biosynthesis , Insulin Resistance , Lipid Metabolism/drug effects , Metabolic Networks and Pathways/drug effects , Oxidative Stress/drug effects
5.
J Proteome Res ; 18(9): 3245-3258, 2019 09 06.
Article in English | MEDLINE | ID: mdl-31317746

ABSTRACT

For the treatment of patients with prediabetes or diabetes, clinical evidence has emerged that ß-cell function can be restored by glucose-lowering therapeutic strategies. However, little is known about the molecular mechanisms underlying this functional adaptive behavior of the pancreatic ß-cell. This study examines the dynamic changes in protein expression and phosphorylation state associated with (pro)insulin production and secretory pathway function mediated by euglycemia to induce ß-cell rest in obese/diabetic db/db islet ß-cells. Unbiased quantitative profiling of the protein expression and phosphorylation events that occur upon ß-cell adaption during the transition from hyperglycemia to euglycemia was assessed in isolated pancreatic islets from obese diabetic db/db and wild-type (WT) mice using quantitative proteomics and phosphoproteomics together with bioinformatics analysis. Dynamic changes in the expression and phosphorylation of proteins associated with pancreatic ß-cell (pro)insulin production and complementary regulated-secretory pathway regulation were observed in obese diabetic db/db islets in a hyperglycemic environment, relative to WT mouse islets in a normal euglycemic environment, that resolved when isolated db/db islets were exposed to euglycemia for 12 h in vitro. By similarly treating WT islets in parallel, the effects of tissue culture could be mostly eliminated and only those changes associated with resolution by euglycemia were assessed. Among such regulated protein phosphorylation-dependent signaling events were those associated with COPII-coated vesicle-dependent ER exit, ER-to-Golgi trafficking, clathrin-coat disassembly, and a particular association for the luminal Golgi protein kinase, FAM20C, in control of distal secretory pathway trafficking, sorting, and granule biogenesis. Protein expression and especially phosphorylation play key roles in the regulation of (pro)insulin production, correlative secretory pathway trafficking, and the restoration of ß-cell secretory capacity in the adaptive functional ß-cell response to metabolic demand, especially that mediated by glucose.


Subject(s)
Calcium-Binding Proteins/genetics , Diabetes Mellitus, Type 2/drug therapy , Extracellular Matrix Proteins/genetics , Prediabetic State/drug therapy , Proteomics , Animals , Blood Glucose/drug effects , COP-Coated Vesicles/genetics , Diabetes Mellitus, Type 2/blood , Disease Models, Animal , Glucose/metabolism , Golgi Apparatus/drug effects , Humans , Hyperglycemia/drug therapy , Hyperglycemia/genetics , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/drug effects , Mice , Mice, Inbred NOD , Obesity/drug therapy , Obesity/genetics , Prediabetic State/blood , Protein Transport/drug effects
6.
Cell Signal ; 42: 127-133, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29042286

ABSTRACT

Inositol pyrophosphates have emerged as important regulators of many critical cellular processes from vesicle trafficking and cytoskeletal rearrangement to telomere length regulation and apoptosis. We have previously demonstrated that 5-di-phosphoinositol pentakisphosphate, IP7, is at a high level in pancreatic ß-cells and is important for insulin exocytosis. To better understand IP7 regulation in ß-cells, we used an insulin secreting cell line, HIT-T15, to screen a number of different pharmacological inhibitors of inositide metabolism for their impact on cellular IP7. Although the inhibitors have diverse targets, they all perturbed IP7 levels. This made us suspicious that indirect, off-target effects of the inhibitors could be involved. It is known that IP7 levels are decreased by metabolic poisons. The fact that the inositol hexakisphosphate kinases (IP6Ks) have a high Km for ATP makes IP7 synthesis potentially vulnerable to ATP depletion. Furthermore, many kinase inhibitors are targeted to the ATP binding site of kinases, but given the similarity of such sites, high specificity is difficult to achieve. Here, we show that IP7 concentrations in HIT-T15 cells were reduced by inhibitors of PI3K (wortmannin, LY294002), PI4K (Phenylarsine Oxide, PAO), PLC (U73122) and the insulin receptor (HNMPA). Each of these inhibitors also decreased the ATP/ADP ratio. Thus reagents that compromise energy metabolism reduce IP7 indirectly. Additionally, PAO, U73122 and LY294002 also directly inhibited the activity of purified IP6K. These data are of particular concern for those studying signal transduction in pancreatic ß-cells, but also highlight the fact that employment of these inhibitors could have erroneously suggested the involvement of key signal transduction pathways in various cellular processes. Conversely, IP7's role in cellular signal transduction is likely to have been underestimated.


Subject(s)
Adenosine Triphosphate/metabolism , Enzyme Inhibitors/pharmacology , Inositol Phosphates/antagonists & inhibitors , Insulin-Secreting Cells/drug effects , Phosphotransferases (Phosphate Group Acceptor)/antagonists & inhibitors , Adenosine Diphosphate/metabolism , Adenosine Triphosphate/antagonists & inhibitors , Androstadienes/pharmacology , Animals , Arsenicals/pharmacology , Cell Line , Chromones/pharmacology , Cricetulus , Estrenes/pharmacology , Gene Expression , Humans , Inositol Phosphates/metabolism , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/metabolism , Morpholines/pharmacology , Phosphotransferases (Phosphate Group Acceptor)/genetics , Phosphotransferases (Phosphate Group Acceptor)/metabolism , Pyrrolidinones/pharmacology , Receptor, Insulin/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Succinimides/pharmacology , Triazoles/pharmacology , Wortmannin
7.
Adv Exp Med Biol ; 975 Pt 1: 319-328, 2017.
Article in English | MEDLINE | ID: mdl-28849466

ABSTRACT

This research aims at figure out the effects and the pathway of taurine on insulin in islet cells cultured in vitro treated by STZ. In the experiment, islet cells were isolated from pancreatic tissue by in situ perfusion with collagenase V. The pancreatic islet cells, maintained in RPMI 1640 culture medium were divided into six groups: C: control, E: supplemented with 10 mmol/L of taurine, group M, T1, T2 and T3 was treated with STZ (0.5 mmol/L), at the same time, taurine were added in group T1,T2 and T3 for 30 min, and then culture medium were collected by centrifugation and then insulin levels were detected by radioimmunoassay, the cells were then rinsed with Hanks, and 0,10, 0, 5, 10, 20 mmol/L of taurine in group C, E, M, T1, T2 and T3 were added for 24 h respectively. Total RNA was extracted, then insulin gene and its transcription regulator such as PDX-1, NeuroD1 were amplified by semi-quantitative RT-PCR. The results showed that, the release of insulin from islet cells treated by STZ could be inhibited by taurine, gene expression of insulin, PDX-1 and NeuroD1 in STZ group decreased significantly, which were up-regulated by taurine administration. In conclusion, taurine exerts a certain degree of protective and reconstructive effects on islet cells treated by STZ.


Subject(s)
Insulin/biosynthesis , Islets of Langerhans/drug effects , Streptozocin/toxicity , Taurine/pharmacology , Animals , Islets of Langerhans/metabolism , Male , Rats , Rats, Wistar
8.
Xenotransplantation ; 24(3)2017 05.
Article in English | MEDLINE | ID: mdl-28397308

ABSTRACT

BACKGROUND: Dead islets replaced with viable islets are a promising offer to restore normal insulin production to a person with diabetes. The main reason for establishing a new islet source for transplantation is the insufficiency of human donor pancreas while using xenogeneic islets perhaps assists this problem. The expression of PDX1 is essential for the pancreas expansion. In mature ß-cells, PDX1 has several critical roles such as glucose sensing, insulin synthesis, and insulin secretion. In this study, we aimed to evaluate the expression of pancreatic duodenal homeobox-1 (PDX1) in treated caprine islets in culture and to assess the protective effects of antioxidant factors on the PDX1 gene in cultured caprine islets. MATERIALS AND METHODS: Purified islets were treated with serum-free, serum, IBMX, tocopherol, or IBMX and tocopherol media. Quantitative polymerase chain reaction and Western blotting were carried out to compare the expression levels of PDX1 in treated purified islets cultured with different media. RESULTS: Islets treated with IBMX/tocopherol exhibited the highest fold change in the relative expression of PDX1 on day 5 post-treatment (relative expression: 6.80±2.08), whereas serum-treated islets showed the lowest fold changes in PDX1 expression on day 5 post-treatment (0.67±0.36), as compared with the expression on day 1 post-treatment. Insulin production and viability tests of purified islets showed superiority of islet at supplemented serum-free media with IBMX/tocopherol compared to other cultures (53.875%±1.59%). CONCLUSIONS: Our results indicated that supplemented serum-free medium with tocopherol and IBMX enhances viability and PDX1 gene expression compared to serum-added and serum-free media.


Subject(s)
Goats/genetics , Goats/physiology , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Islets of Langerhans/physiology , Trans-Activators/genetics , Trans-Activators/physiology , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Antioxidants/pharmacology , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Culture Media , Culture Media, Serum-Free , Gene Expression/drug effects , Genes, Homeobox , In Vitro Techniques , Insulin/biosynthesis , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Male , Tocopherols/pharmacology
9.
Clin Sci (Lond) ; 131(8): 673-687, 2017 Apr 25.
Article in English | MEDLINE | ID: mdl-28188238

ABSTRACT

The aim of the present study was to demonstrate the role of autophagy and incretins in the fructose-induced alteration of ß-cell mass and function. Normal Wistar rats were fed (3 weeks) with a commercial diet without (C) or with 10% fructose in drinking water (F) alone or plus sitagliptin (CS and FS) or exendin-4 (CE and FE). Serum levels of metabolic/endocrine parameters, ß-cell mass, morphology/ultrastructure and apoptosis, vacuole membrane protein 1 (VMP1) expression and glucose-stimulated insulin secretion (GSIS) were studied. Complementary to this, islets isolated from normal rats were cultured (3 days) without (C) or with F and F + exendin-4 or chloroquine. Expression of autophagy-related proteins [VMP1 and microtubule-associated protein light chain 3 (LC3)], apoptotic/antiapoptotic markers (caspase-3 and Bcl-2), GSIS and insulin mRNA levels were measured. F rats developed impaired glucose tolerance (IGT) and a significant increase in plasma triacylglycerols, thiobarbituric acid-reactive substances, insulin levels, homoeostasis model assessment (HOMA) for insulin resistance (HOMA-IR) and ß-cell function (HOMA-ß) indices. A significant reduction in ß-cell mass was associated with an increased apoptotic rate and morphological/ultrastructural changes indicative of autophagic activity. All these changes were prevented by either sitagliptin or exendin-4. In cultured islets, F significantly enhanced insulin mRNA and GSIS, decreased Bcl-2 mRNA levels and increased caspase-3 expression. Chloroquine reduced these changes, suggesting the participation of autophagy in this process. Indeed, F induced the increase of both VMP1 expression and LC3-II, suggesting that VMP1-related autophagy is activated in injured ß-cells. Exendin-4 prevented islet-cell damage and autophagy development. VMP1-related autophagy is a reactive process against F-induced islet dysfunction, being prevented by exendin-4 treatment. This knowledge could help in the use of autophagy as a potential target for preventing progression from IGT to type 2 diabetes mellitus.


Subject(s)
Autophagy/drug effects , Diet/adverse effects , Fructose/pharmacology , Incretins/pharmacology , Insulin-Secreting Cells/drug effects , Membrane Proteins/physiology , Animals , Autophagy/physiology , Body Weight , Cells, Cultured , Drug Evaluation, Preclinical/methods , Energy Intake , Exenatide , Fructose/administration & dosage , Glucose Intolerance/etiology , Glucose Intolerance/pathology , Glucose Intolerance/prevention & control , Glucose Tolerance Test , Hypoglycemic Agents/pharmacology , Insulin/biosynthesis , Insulin/genetics , Insulin-Secreting Cells/ultrastructure , Male , Microscopy, Electron , Peptides/pharmacology , RNA, Messenger/genetics , Rats, Wistar , Sitagliptin Phosphate/pharmacology , Venoms/pharmacology
10.
PLoS One ; 11(1): e0147391, 2016.
Article in English | MEDLINE | ID: mdl-26807719

ABSTRACT

The novel sodium glucose co-transporter 2 (SGLT2) inhibitor empagliflozin has recently been reported to improve glycemic control in streptozotocin-induced type 1 diabetic rats in an insulin-independent manner, via an increase in urinary glucose output. We investigated the potential of empagliflozin to recover insulin pathways in type 1 diabetes by improving pancreatic ß-cell mass. Blood glucose homeostasis was assessed by an intraperitoneal glucose tolerance test. Serum insulin levels and insulin mRNA expression were determined using commercial insulin ELISA kits and real-time quantitative polymerase chain reaction, respectively. Immunohistochemistry was used to investigate ß-cell areas, ß-cell proliferation, apoptosis of pancreatic ß-cells, and reactive oxygen species production in the pancreatic ß-cells. Results showed that glucose tolerance was significantly improved in streptozotocin-induced type 1 diabetic mice treated with empagliflozin. Empagliflozin-treated mice also showed an increase in insulin mRNA expression. Higher serum insulin levels were detected in mice treated with empagliflozin compared with the vehicle group. Immunohistochemistry indicated that ß-cell area/total pancreatic area and the expression of cell proliferation marker Ki-67 (co-stained with insulin) were significantly enhanced by empagliflozin treatment. These effects were due, probably, to a reduction in apoptosis and reactive oxygen species in the pancreatic ß-cells. Taken together, the results of this study indicate that empagliflozin may have a beneficial effect on preserving ß-cell regeneration, thus improving blood glucose homeostasis in type 1 diabetes mellitus, probably via the protection of pancreatic ß-cell from glucotoxicity-induced oxidative stress.


Subject(s)
Benzhydryl Compounds/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Glucose/metabolism , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Insulin-Secreting Cells/drug effects , Animals , Apoptosis/drug effects , Area Under Curve , Benzhydryl Compounds/administration & dosage , Benzhydryl Compounds/therapeutic use , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 1/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Glucose Tolerance Test , Glucosides/administration & dosage , Glucosides/therapeutic use , Homeostasis , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/therapeutic use , Insulin/biosynthesis , Insulin/blood , Insulin/genetics , Insulin-Secreting Cells/pathology , Male , Mice , Mice, Inbred C57BL , Organ Size/drug effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Sodium-Glucose Transporter 2 , Sodium-Glucose Transporter 2 Inhibitors
11.
Oncotarget ; 6(31): 30568-78, 2015 Oct 13.
Article in English | MEDLINE | ID: mdl-26375250

ABSTRACT

In this study, we tested whether a standardized epigallocatechin-3-gallate (EGCG) rich green tea extract (comprising > 90% EGCG) affects fitness and lifespan as well as parameters of glucose metabolism and energy homeostasis in the fruit fly, Drosophila melanogaster. Following the application of the green tea extract a significant increase in the mean lifespan (+ 3.3 days) and the 50% survival (+ 4.3 days) as well as improved fitness was detected. These effects went along an increased expression of Spargel, the homolog of mammalian PGC1α, which has been reported to affect lifespan in flies. Intriguingly, in flies, treatment with the green tea extract decreased glucose concentrations, which were accompanied by an inhibition of α-amylase and α-glucosidase activity. Computational docking analysis proved the potential of EGCG to dock into the substrate binding pocket of α-amylase and to a greater extent into α-glucosidase. Furthermore, we demonstrate that EGCG downregulates insulin-like peptide 5 and phosphoenolpyruvate carboxykinase, major regulators of glucose metabolism, as well as the Drosophila homolog of leptin, unpaired 2. We propose that a decrease in glucose metabolism in connection with an upregulated expression of Spargel contribute to the better fitness and the extended lifespan in EGCG-treated flies.


Subject(s)
Antioxidants/pharmacology , Catechin/analogs & derivatives , Energy Metabolism/drug effects , Glucose/metabolism , Longevity/physiology , Animals , Camellia sinensis/metabolism , Catechin/pharmacology , Drosophila Proteins/biosynthesis , Drosophila melanogaster/metabolism , Homeostasis/drug effects , Insulin/biosynthesis , Longevity/drug effects , Molecular Docking Simulation , Phosphoenolpyruvate Carboxykinase (ATP)/biosynthesis , Plant Extracts/pharmacology , Positive Transcriptional Elongation Factor B/biosynthesis , Proteins , Transcription Factors/biosynthesis , Up-Regulation , alpha-Amylases/metabolism , alpha-Glucosidases/metabolism
12.
J Psychopharmacol ; 29(12): 1271-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26349558

ABSTRACT

Meal-induced insulin sensitization (MIS), an endogenous adaptive mechanism is activated post-prandially. Reduced MIS leads to diabetes, but its activation improves insulin sensitivity. MIS is preserved to single olanzapine administration, therefore we aimed to investigate the chronic effect of olanzapine on fasted-state insulin sensitivity and on MIS in female Sprague-Dawley rats. Daily food and water intake, stool and urine production and body weight were determined. The MIS was characterized by a rapid insulin sensitivity test. Fasting hepatic and peripheral insulin sensitivity were determined by a hyperinsulinaemic euglycaemic glucose clamping supplemented with radiotracer technique. Fasted and post-prandial blood samples were obtained for plasma insulin, leptin, ghrelin, amylin, GLP-1, GIP, PYY and PP determination. Adiposity was characterized by weighing intra-abdominal and inguinal fat pads. Olanzapine caused hepatic insulin resistance and a reduced metabolic clearance rate of insulin, but the MIS retained its function. Body weight and adiposity were enhanced, but olanzapine failed to increase food intake. Fasting insulin and leptin were elevated and the post-prandial reduction in ghrelin level was inhibited by olanzapine.The MIS remained functionally intact after long-term olanzapine treatment. Altered insulin, leptin and ghrelin levels indicate olanzapine-induced metabolic derangements. Pharmacological activation of MIS could potentially be exploited to treat or prevent olanzapine-induced insulin resistance.


Subject(s)
Benzodiazepines/administration & dosage , Gastrointestinal Hormones/blood , Insulin Resistance/physiology , Insulin/biosynthesis , Animals , Blood Glucose/drug effects , Body Weight/drug effects , Eating/drug effects , Female , Ghrelin/blood , Leptin/blood , Obesity/blood , Olanzapine , Rats , Rats, Sprague-Dawley
13.
Mol Med Rep ; 12(2): 2547-54, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25955179

ABSTRACT

Previously, powdered persimmon leaves have been reported to have glucose- and lipid-lowering effects in diabetic (db/db) mice. As persimmon leaf is commonly consumed as tea, an aqueous extract of persimmon leaves (PLE) was prepared and its anti-diabetic efficacy was investigated. In the present study, PLE was tested for its inhibitory activity on α-glucosidase in vitro. An oral maltose tolerance test was performed in diabetic mice. Next, the acute effect of PLE was examined in streptozotocin-induced diabetic mice. Last, the long-term effect of PLE supplementation was assessed in db/db after eight weeks. An oral glucose tolerance test, biochemical parameters, as well as histological analyses of liver and pancreas were evaluated at the end of the study. PLE inhibited α-glucosidase activity and increased antioxidant capacity. Streptozotocin-induced diabetic mice pre-treated with PLE displayed hypoglycemic activity. Daily oral supplementation with PLE for eight weeks reduced body weight gain without affecting food intake, enhanced the glucose tolerance during the oral glucose tolerance test (OGTT), improved blood lipid parameters, suppressed fat accumulation in the liver and maintained islet structure in db/db mice. Further mechanistic study showed that PLE protected pancreatic islets from glucotoxicity. In conclusion, the results of the present study indicated that PLE exhibits considerable anti-diabetic effects through α-glucosidase inhibition and through the maintenance of functional ß-cells. These results provided a rationale for the use of persimmon leaf tea for the maintenance of normal blood glucose levels in diabetic patients.


Subject(s)
Antioxidants/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diospyros/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Hypoglycemic Agents/pharmacology , Phytotherapy/methods , Animals , Antioxidants/isolation & purification , Biphenyl Compounds/antagonists & inhibitors , Blood Glucose/drug effects , Body Weight/drug effects , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Eating , Glucose Tolerance Test , Glycoside Hydrolase Inhibitors/isolation & purification , Humans , Hypoglycemic Agents/isolation & purification , Insulin/agonists , Insulin/biosynthesis , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Lipogenesis/drug effects , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Picrates/antagonists & inhibitors , Plant Extracts/chemistry , Plant Leaves/chemistry , Streptozocin , alpha-Glucosidases/metabolism
14.
PLoS One ; 9(10): e109607, 2014.
Article in English | MEDLINE | ID: mdl-25299668

ABSTRACT

OBJECTIVES: To examine whether combined vitamin D and calcium supplementation improves insulin sensitivity, insulin secretion, ß-cell function, inflammation and metabolic markers. DESIGN: 6-month randomized, placebo-controlled trial. PARTICIPANTS: Ninety-five adults with serum 25-hydroxyvitamin D [25(OH)D] ≤55 nmol/L at risk of type 2 diabetes (with prediabetes or an AUSDRISK score ≥15) were randomized. Analyses included participants who completed the baseline and final visits (treatment n = 35; placebo n = 45). INTERVENTION: Daily calcium carbonate (1,200 mg) and cholecalciferol [2,000-6,000 IU to target 25(OH)D >75 nmol/L] or matching placebos for 6 months. MEASUREMENTS: Insulin sensitivity (HOMA2%S, Matsuda index), insulin secretion (insulinogenic index, area under the curve (AUC) for C-peptide) and ß-cell function (Matsuda index x AUC for C-peptide) derived from a 75 g 2-h OGTT; anthropometry; blood pressure; lipid profile; hs-CRP; TNF-α; IL-6; adiponectin; total and undercarboxylated osteocalcin. RESULTS: Participants were middle-aged adults (mean age 54 years; 69% Europid) at risk of type 2 diabetes (48% with prediabetes). Compliance was >80% for calcium and vitamin D. Mean serum 25(OH)D concentration increased from 48 to 95 nmol/L in the treatment group (91% achieved >75 nmol/L), but remained unchanged in controls. There were no significant changes in insulin sensitivity, insulin secretion and ß-cell function, or in inflammatory and metabolic markers between or within the groups, before or after adjustment for potential confounders including waist circumference and season of recruitment. In a post hoc analysis restricted to participants with prediabetes, a significant beneficial effect of vitamin D and calcium supplementation on insulin sensitivity (HOMA%S and Matsuda) was observed. CONCLUSIONS: Daily vitamin D and calcium supplementation for 6 months may not change OGTT-derived measures of insulin sensitivity, insulin secretion and ß-cell function in multi-ethnic adults with low vitamin D status at risk of type 2 diabetes. However, in participants with prediabetes, supplementation with vitamin D and calcium may improve insulin sensitivity. TRIAL REGISTRATION: Australian New Zealand Clinical Trials Registry ACTRN12609000043235.


Subject(s)
Calcium, Dietary/administration & dosage , Cholecalciferol/administration & dosage , Diabetes Mellitus, Type 2/prevention & control , Dietary Supplements , Prediabetic State/diet therapy , Vitamin D Deficiency/diet therapy , Adiponectin/metabolism , Adult , Aged , Blood Glucose/metabolism , C-Peptide/biosynthesis , C-Reactive Protein/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Female , Humans , Insulin/biosynthesis , Insulin/pharmacology , Insulin Resistance , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Interleukin-6/metabolism , Male , Middle Aged , Osteocalcin/metabolism , Pilot Projects , Prediabetic State/metabolism , Prediabetic State/physiopathology , Tumor Necrosis Factor-alpha/metabolism , Vitamin D Deficiency/metabolism , Vitamin D Deficiency/physiopathology
15.
Bone ; 65: 42-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24815919

ABSTRACT

Maximizing peak bone mass is an important factor in osteoporosis prevention. Resistance exercise increases bone mass and strength, while nutritional supplements have beneficial effects on bone loss reduction. We have previously shown that the combined intake of sucrose and amino acids (AA), which is strongly insulinogenic, efficiently increased muscle protein synthesis. To investigate the effects of sugar and an AA solution immediately after resistance exercise, we compared insulinogenic sucrose and non-insulinogenic fructose combined with an AA solution with or without resistance exercise. Sucrose intake immediately after resistance exercise increased the trabecular bone mass and compressive maximum load compared with fructose+AA intake after exercise. Additionally, combined sucrose+AA and exercise increased trabecular bone formation and decreased bone resorption more than combined fructose and exercise. Serum insulin levels were greatly increased by sucrose+AA intake with exercise. In culture experiments, neither sugar+AA affected osteoblast and osteoclast differentiation. In a gene expression study, sucrose+AA intake after resistance exercise was shown to upregulate the Runx2 expression level and decrease RANKL/OPG ratio. These results suggest that the combined intake of sucrose and an AA solution immediately after resistance exercise exerts anabolic effects on bone by altering gene expression related to bone remodeling. Although translation of our bone remodeling findings from animal to human studies has been challenging, our findings suggest that exercise with sugar+AA intake may contribute to improved bone health.


Subject(s)
Amino Acids/administration & dosage , Bone and Bones/physiology , Fructose/administration & dosage , Insulin/biosynthesis , Physical Conditioning, Animal , Sucrose/administration & dosage , 3T3 Cells , Absorptiometry, Photon , Animals , Base Sequence , Bone Density , DNA Primers , Mice , Rats , Real-Time Polymerase Chain Reaction
16.
J Diabetes Res ; 2014: 862473, 2014.
Article in English | MEDLINE | ID: mdl-24812635

ABSTRACT

In China, TianMai Xiaoke tablet (TM) is used to treat type 2 diabetes. However, the exact mechanism of TM is not clear. This study is to investigate the effect of TM on glucose metabolism in diabetic rats and to identify whether TM takes a direct action through microRNAs on islet. Rats were divided into control group, diabetic group, low dose of TM group (TML), and high dose of TM group (TMH). Pancreas samples were analyzed using microRNA array and Q-PCR. Eight-week treatment with TM significantly decreased fasting blood glucose. The blood glucose was significantly reduced in TM-treated groups before and after oral glucose administration. Fasting insulin and HOMA-IR were suppressed in TM-treated groups. miR-448, let-7b, miR-540, miR-296, miR-880, miR-200a, miR-500, miR-10b, miR-336, miR-30d, miR-208, let-7e, miR-142-5p, miR-874, miR-375, miR-879, miR-501, and miR-188 were upregulated, while miR-301b, miR-134, and miR-652 were downregulated in TMH group. Through target gene analysis and real-time PCR verification, we found that these miRNAs, especially miR-375 and miR-30d, can stimulate insulin secretion in islet. Our data suggest that TM can improve blood glucose in diabetic rats which involved increasing the expression of miR-375 and miR-30d to activate insulin synthesis in islet.


Subject(s)
Chromium/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Drugs, Chinese Herbal/therapeutic use , Hypoglycemic Agents/therapeutic use , Islets of Langerhans/drug effects , MicroRNAs/metabolism , Animals , Body Weight/drug effects , Chromium/administration & dosage , Chromium/adverse effects , Chromium Compounds/administration & dosage , Chromium Compounds/adverse effects , Chromium Compounds/therapeutic use , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/adverse effects , Hyperglycemia/prevention & control , Hyperinsulinism/prevention & control , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Insulin/biosynthesis , Insulin/metabolism , Insulin Resistance , Insulin Secretion , Islets of Langerhans/metabolism , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Up-Regulation/drug effects
17.
Curr Alzheimer Res ; 11(2): 119-27, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24359498

ABSTRACT

Increasing evidence suggests that dysregulation of brain insulin/insulin receptor (InsR) and insulin signaling cascade are associated with the pathogenesis of Alzheimer's disease (AD). Our group has designed and synthesized a series of multi-target iron chelating, brain permeable compounds for AD. One leading multi-target compound, M30 possesses the neuroprotective N-propargyl moiety of the anti-Parkinsonian, monoamine oxidase (MAO)-B inhibitor, rasagiline (Azilect®) and the antioxidant-iron chelating moiety of an 8-hydroxyquinoline derivative of the iron chelator, VK28. Positive outcomes for the behavioral/cognitive and neuroprotective effects of M30 were recently obtained in preclinical experimental studies, regarding pathological aspects relevant to ageing and AD. We report that chronic treatment with M30 (1 and 5 mg/kg p.o; three times a week for 9 months) significantly elevated cortical insulin and InsR transcript and protein expression, respectively and increased the phosphorylated form of glycogen synthase kinase-3ß in the frontal cortex of amyloid precursor protein (APP) and presenilin 1 (PS1) double transgenic mice. In addition, M30 treatment upregulated the levels of hypoxia-inducible factor (HIF)-1α and expression of its target genes involved in glycolysis including, aldolase A, enolase-1 and glucose transporter-1 (Glut-1), in the frontal cortex of APP/PS1 mice. Treatment with M30 also lead to an increase in the hepatic protein expression levels of InsR and Glut-1 and lowered the increase in blood glucose levels following glucose tolerance test. The present findings indicate that the multifunctional iron chelating drug, M30 regulates major brain glucose metabolism parameters and thus, might be beneficial for AD, in which impaired neuronal insulin signaling and Glut expression have been implicated.


Subject(s)
Alzheimer Disease/metabolism , Glucose Transporter Type 1/biosynthesis , Hydroxyquinolines/administration & dosage , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Insulin/biosynthesis , Iron Chelating Agents/administration & dosage , Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Animals , Drug Delivery Systems/methods , Frontal Lobe/drug effects , Frontal Lobe/metabolism , Glycolysis/drug effects , Glycolysis/physiology , Male , Mice , Presenilin-1/genetics , Signal Transduction/drug effects , Signal Transduction/physiology
18.
Free Radic Biol Med ; 65: 1548-1556, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23867154

ABSTRACT

Whereas selenium was found to act as an insulin mimic and to be antidiabetic in earlier studies, recent animal experiments and human trials have shown an unexpected risk of prolonged high Se intake in potentiating insulin resistance and type 2 diabetes. Elevating dietary Se intake (0.4 to 3.0mg/kg of diet) above the nutrient requirements, similar to overproduction of selenoproteins, led to insulin resistance and/or diabetes-like phenotypes in mice, rats, and pigs. Although its diabetogenic mechanism remains unclear, high Se intake elevated activity or production of selenoproteins including GPx1, MsrB1, SelS, and SelP. This upregulation diminished intracellular reactive oxygen species and then dysregulated key regulators of ß cells and insulin synthesis and secretion, leading to chronic hyperinsulinemia. Overscavenging intracellular H2O2 also attenuated oxidative inhibition of protein tyrosine phosphatases and suppressed insulin signaling. High Se intake might affect expression and/or function of key regulators of glycolysis, gluconeogenesis, and lipogenesis. Future research is needed to find out if certain forms of Se metabolites in addition to selenoproteins and if mechanisms other than intracellular redox control mediate the diabetogenic effects of high Se intake. Furthermore, a potential interactive role of high Se intake in the interphase of carcinogenesis and diabetogenesis should be explored to make optimal use of Se in human nutrition and health.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Hyperinsulinism , Insulin Resistance , Selenium/pharmacology , Animals , Gluconeogenesis/drug effects , Glycolysis/drug effects , Hydrogen Peroxide/metabolism , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Lipogenesis/drug effects , Mice , Oxidation-Reduction/drug effects , Rats , Selenoproteins/metabolism , Signal Transduction , Swine
19.
Nutr Res Rev ; 26(1): 1-11, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23286442

ABSTRACT

Zn is an essential trace element, involved in many different cellular processes. A relationship between Zn, pancreatic function and diabetes was suggested almost 70 years ago. To emphasise the importance of Zn in biology, the history of Zn research in the field of diabetes along with a general description of Zn transporter families will be reviewed. The paper will then focus on the effects of Zn on pancreatic ß-cell function, including insulin synthesis and secretion, Zn signalling in the pancreatic islet, the redox functions of Zn and its target genes. The recent association of two 'Zn genes', i.e. metallothionein (MT) and Zn transporter 8 (SLC 30A8), with type 2 diabetes at the genetic level and with insulin secretion in clinical studies offers a potential new way to identify new drug targets to modulate Zn homeostasis directly in ß-cells. The action of Zn for insulin action in its target organs, as Zn signalling in other pancreatic islet cells, will be addressed. Therapeutic Zn-insulin preparations and the influence of Zn and Zn transporters in type 1 diabetes will also be discussed. An extensive review of the literature on the clinical studies using Zn supplementation in the prevention and treatment of both types of diabetes, including complications of the disease, will evaluate the overall beneficial effects of Zn supplementation on blood glucose control, suggesting that Zn might be a candidate ion for diabetes prevention and therapy. Clearly, the story of the links between Zn, pancreatic islet cells and diabetes is only now unfolding, and we are presently only at the first chapter.


Subject(s)
Diabetes Mellitus/physiopathology , Islets of Langerhans/physiopathology , Zinc/physiology , Carrier Proteins/physiology , Diabetes Mellitus/prevention & control , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 1/prevention & control , Diabetes Mellitus, Type 2/physiopathology , Diabetes Mellitus, Type 2/prevention & control , Dietary Supplements , Humans , Insulin/biosynthesis , Insulin/metabolism , Insulin Secretion , Metallothionein/physiology , Signal Transduction , Zinc/administration & dosage
20.
Phytother Res ; 27(7): 1006-11, 2013 Jul.
Article in English | MEDLINE | ID: mdl-22911568

ABSTRACT

Herbal medicines, especially plant-derived extracts, have been used to treat Type 2 diabetes mellitus (T2DM) for many centuries, and offer the potential of cheap and readily available alternatives to conventional pharmaceuticals in developing countries. Extracts of Gymnema sylvestre (GS) have anti-diabetic activities and have been used as a folk medicine in India for centuries. We have investigated the effects of a novel high molecular weight GS extract termed OSA® on glucose tolerance in insulin-resistant ob/ob mice, and on insulin secretion and synthesis by isolated mouse islets. Single administration of OSA® (500 mg/kg) to ob/ob mice 30 min before an intraperitoneal glucose load improved their abnormal glucose tolerance. In vitro studies indicated that OSA® (0.25 mg/ml) initiated rapid and reversible increases in insulin secretion from isolated mouse islets at substimulatory (2 mM) and stimulatory (20 mM) glucose concentrations. In addition, prolonged treatment (24-48 h) of mouse islets with OSA® elevated the expression of preproinsulin mRNA and maintained the total insulin content of mouse islets in the presence of stimulated insulin secretion. These effects of OSA® are consistent with its potential use as a therapy for the hyperglycemia associated with obesity-related T2DM.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucose Intolerance/drug therapy , Gymnema sylvestre/chemistry , Hypoglycemic Agents/therapeutic use , Insulin/biosynthesis , Islets of Langerhans/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Female , Insulin/genetics , Insulin/metabolism , Insulin Resistance , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/metabolism , Protein Precursors/genetics , Protein Precursors/metabolism , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL