Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Diabetologia ; 59(7): 1533-1541, 2016 07.
Article in English | MEDLINE | ID: mdl-27053237

ABSTRACT

AIMS/HYPOTHESIS: The action of incretin hormones including glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) is potentiated in animal models defective in glucagon action. It has been reported that such animal models maintain normoglycaemia under streptozotocin (STZ)-induced beta cell damage. However, the role of GIP in regulation of glucose metabolism under a combination of glucagon deficiency and STZ-induced beta cell damage has not been fully explored. METHODS: In this study, we investigated glucose metabolism in mice deficient in proglucagon-derived peptides (PGDPs)-namely glucagon gene knockout (GcgKO) mice-administered with STZ. Single high-dose STZ (200 mg/kg, hSTZ) or moderate-dose STZ for five consecutive days (50 mg/kg × 5, mSTZ) was administered to GcgKO mice. The contribution of GIP to glucose metabolism in GcgKO mice was also investigated by experiments employing dipeptidyl peptidase IV (DPP4) inhibitor (DPP4i) or Gcg-Gipr double knockout (DKO) mice. RESULTS: GcgKO mice developed severe diabetes by hSTZ administration despite the absence of glucagon. Administration of mSTZ decreased pancreatic insulin content to 18.8 ± 3.4 (%) in GcgKO mice, but ad libitum-fed blood glucose levels did not significantly increase. Glucose-induced insulin secretion was marginally impaired in mSTZ-treated GcgKO mice but was abolished in mSTZ-treated DKO mice. Although GcgKO mice lack GLP-1, treatment with DPP4i potentiated glucose-induced insulin secretion and ameliorated glucose intolerance in mSTZ-treated GcgKO mice, but did not increase beta cell area or significantly reduce apoptotic cells in islets. CONCLUSIONS/INTERPRETATION: These results indicate that GIP has the potential to ameliorate glucose intolerance even under STZ-induced beta cell damage by increasing insulin secretion rather than by promoting beta cell survival.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Insulin/metabolism , Proglucagon/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Insulin-Secreting Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Proglucagon/deficiency , Streptozocin/toxicity
2.
J Diabetes Complications ; 28(5): 588-95, 2014.
Article in English | MEDLINE | ID: mdl-25027388

ABSTRACT

AIMS/HYPOTHESIS: Although the initial healing stage involves a re-epithelialization in humans, diabetic foot ulceration (DFU) has been investigated using rodent models with wounds on the thigh skin, in which a wound contraction is initiated. In this study, we established a rodent model of DFU on the plantar skin and evaluated the therapeutic efficacy of bone-marrow-derived mesenchymal stem cells (BM-MSCs) in this model. METHODS: The wounds made on the hind paws or thighs of streptozotocin induced diabetic or control rats were treated with BM-MSCs. Expression levels of phosphorylated focal adhesion kinase (pFAK), matrix metaroprotease (MMP)-2, EGF, and IGF-1, were evaluated in human keratinocytes, which were cultured in conditioned media of BM-MSCs (MSC-CM) with high glucose levels. RESULTS: Re-epithelialization initiated the healing process on the plantar, but not on the thigh, skin. The therapy utilizing BM-MSCs ameliorated the delayed healing in diabetic rats. In the keratinocytes cultured with MSC-CM, the decreased pFAK levels in the high glucose condition were restored, and the MMP2, EGF, and IGF-1 levels increased. CONCLUSIONS/INTERPRETATION: Our study established a novel rat DFU model. The impaired healing process in diabetic rats was ameliorated by transplantation of BM-MSCs. This amelioration might be accounted for by the modification of keratinocyte functions.


Subject(s)
Diabetes Mellitus, Experimental , Diabetic Foot/physiopathology , Diabetic Foot/therapy , Keratinocytes/physiology , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Wound Healing , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Foot , Humans , Keratinocytes/pathology , Male , Rats , Rats, Sprague-Dawley , Skin/pathology , Skin/physiopathology , Streptozocin
3.
Obesity (Silver Spring) ; 22(2): 371-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23804363

ABSTRACT

OBJECTIVE: The S100 calcium binding protein B (S100B) implicated in brain inflammation acts via the receptor of advanced glycation end products (RAGE) and is also secreted from adipocytes. We investigated the role of S100B in the interaction between adipocytes and macrophages using a cell-culture model. DESIGN AND METHODS: RAW264.7 macrophages (RAW) were stimulated by recombinant S100B to observe alterations in TNF-α and M1 markers; 3T3-L1 adipocytes (L1) were stimulated by TNF-α to examine S100B secretion. RAW and L1 were then mutually stimulated with conditioned media of each other, or co-cultured. The effects of S100B silencing or a RAGE-neutralizing antibody were also investigated. RESULTS: S100B upregulated TNF-α and M1 markers in RAW, and TNF-α augmented S100B secretion from L1. L1 conditioned media stimulated TNF-α secretion from RAW, and RAW conditioned media increased S100B secretion from L1. The co-culture of RAW and L1 increased TNF-α, S100B, and the expression of M1 markers and the MCP-1 receptor CCR2. The silencing of S100B or RAGE neutralization significantly ameliorated TNF-α hypersecretion from RAW that were stimulated with L1 conditioned media. CONCLUSIONS: Thus, S100B as an adipokine may play a role in the interaction between adipocytes and macrophages to establish a vicious paracrine loop.


Subject(s)
Adipocytes, White/metabolism , Cell Communication , Macrophages/metabolism , Receptors, Immunologic/agonists , S100 Calcium Binding Protein beta Subunit/metabolism , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism , 3T3-L1 Cells , Adipocytes, White/drug effects , Adipocytes, White/immunology , Adipokines/antagonists & inhibitors , Adipokines/genetics , Adipokines/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Cell Communication/drug effects , Cell Line, Transformed , Coculture Techniques , Culture Media, Conditioned/metabolism , Gene Silencing , Immunomodulation/drug effects , Insulin Resistance , Macrophage Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Mice , Obesity/immunology , Obesity/metabolism , Paracrine Communication/drug effects , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Receptors, Immunologic/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , S100 Calcium Binding Protein beta Subunit/antagonists & inhibitors , S100 Calcium Binding Protein beta Subunit/genetics , Signal Transduction/drug effects , Tumor Necrosis Factor-alpha/agonists
4.
Diabetes ; 62(2): 510-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23099862

ABSTRACT

Glucagon and glucagon-like peptide-1 (GLP-1) are produced in pancreatic α-cells and enteroendocrine L-cells, respectively, in a tissue-specific manner from the same precursor, proglucagon, that is encoded by glucagon gene (Gcg), and play critical roles in glucose homeostasis. Here, we studied glucose homeostasis and ß-cell function of Gcg-deficient mice that are homozygous for a Gcg-GFP knock-in allele (Gcg(gfp/gfp)). The Gcg(gfp/gfp) mice displayed improved glucose tolerance and enhanced insulin secretion, as assessed by both oral glucose tolerance test (OGTT) and intraperitoneal glucose tolerance test (IPGTT). Responses of glucose-dependent insulinotropic polypeptide (GIP) to both oral and intraperitoneal glucose loads were unexpectedly enhanced in Gcg(gfp/gfp) mice, and immunohistochemistry localized GIP to pancreatic ß-cells of Gcg(gfp/gfp) mice. Furthermore, secretion of GIP in response to glucose was detected in isolated islets of Gcg(gfp/gfp) mice. Blockade of GIP action in vitro and in vivo by cAMP antagonism and genetic deletion of the GIP receptor, respectively, almost completely abrogated enhanced insulin secretion in Gcg(gfp/gfp) mice. These results indicate that ectopic GIP expression in ß-cells maintains insulin secretion in the absence of proglucagon-derived peptides (PGDPs), revealing a novel compensatory mechanism for sustaining incretin hormone action in islets.


Subject(s)
Gastric Inhibitory Polypeptide/biosynthesis , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Peptide Fragments/metabolism , Proglucagon/metabolism , Animals , Cyclic AMP/antagonists & inhibitors , Gastric Inhibitory Polypeptide/genetics , Gene Deletion , Gene Knock-In Techniques , Glucagon-Like Peptide-1 Receptor , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Glucose Tolerance Test , Homeostasis/genetics , Homeostasis/physiology , Immunohistochemistry , Incretins/metabolism , Insulin Secretion , Insulin-Secreting Cells/cytology , Male , Mice , Proglucagon/analysis , Receptors, Gastrointestinal Hormone/genetics , Receptors, Glucagon/metabolism
5.
Diabetes ; 61(1): 74-84, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22187375

ABSTRACT

Glucagon is believed to be one of the most important peptides for upregulating blood glucose levels. However, homozygous glucagon-green fluorescent protein (gfp) knock-in mice (Gcg(gfp/gfp): GCGKO) are normoglycemic despite the absence of proglucagon-derived peptides, including glucagon. To characterize metabolism in the GCGKO mice, we analyzed gene expression and metabolome in the liver. The expression of genes encoding rate-limiting enzymes for gluconeogenesis was only marginally altered. On the other hand, genes encoding enzymes involved in conversion of amino acids to metabolites available for the tricarboxylic acid cycle and/or gluconeogenesis showed lower expression in the GCGKO liver. The expression of genes involved in the metabolism of fatty acids and nicotinamide was also altered. Concentrations of the metabolites in the GCGKO liver were altered in manners concordant with alteration in the gene expression patterns, and the plasma concentrations of amino acids were elevated in the GCGKO mice. The insulin concentration in serum and phosphorylation of Akt protein kinase in liver were reduced in GCGKO mice. These results indicated that proglucagon-derived peptides should play important roles in regulating various metabolic pathways, especially that of amino acids. Serum insulin concentration is lowered to compensate the impacts of absent proglucagon-derived peptide on glucose metabolism. On the other hand, impacts on other metabolic pathways are only partially compensated by reduced insulin action.


Subject(s)
Amino Acids/blood , Liver/metabolism , Metabolic Diseases/metabolism , Proglucagon/deficiency , Proglucagon/genetics , Amino Acids/metabolism , Animals , Gene Expression Regulation, Enzymologic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver/enzymology , Male , Metabolic Diseases/blood , Metabolic Diseases/genetics , Metabolic Networks and Pathways/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Peptide Fragments/chemistry , Peptide Fragments/deficiency , Peptide Fragments/genetics , Peptide Fragments/metabolism , Proglucagon/chemistry , Proglucagon/metabolism , Up-Regulation
6.
J Diabetes Investig ; 3(5): 432-40, 2012 Oct 18.
Article in English | MEDLINE | ID: mdl-24843603

ABSTRACT

UNLABELLED: Aims/Introduction: Excessive intake of sucrose can cause severe health issues, such as diabetes mellitus. In animal studies, consumption of a high-sucrose diet (SUC) has been shown to cause obesity, insulin resistance and glucose intolerance. However, several in vivo experiments have been carried out using diets with much higher sucrose contents (50-70% of the total calories) than are typically ingested by humans. In the present study, we examined the effects of a moderate SUC on glucose metabolism and the underlying mechanism. MATERIALS AND METHODS: C57BL/6J mice received a SUC (38.5% sucrose), a high-starch diet (ST) or a control diet for 5 weeks. We assessed glucose tolerance, incretin secretion and liver glucose metabolism. RESULTS: An oral glucose tolerance test (OGTT) showed that plasma glucose levels in the early phase were significantly higher in SUC-fed mice than in ST-fed or control mice, with no change in plasma insulin levels at any stage. SUC-fed mice showed a significant improvement in insulin sensitivity. Glucagon-like peptide-1 (GLP-1) secretion 15 min after oral glucose administration was significantly lower in SUC-fed mice than in ST-fed or control mice. Hepatic glucokinase (GCK) activity was significantly reduced in SUC-fed mice. During the OGTT, the accumulation of glycogen in the liver was suppressed in SUC-fed mice in a time-dependent manner. CONCLUSIONS: These results indicate that mice that consume a moderate SUC show glucose intolerance with a reduction in hepatic GCK activity and impairment in GLP-1 secretion. (J Diabetes Invest, doi: 10.1111/j.2040-1124.2012.00208.x, 2012).

7.
Diabetes ; 60(9): 2397-406, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21810596

ABSTRACT

OBJECTIVE: The therapeutic potential of exendin-4, an agonist of the glucagon-like peptide-1 receptor (GLP-1R), on diabetic polyneuropathy (DPN) in streptozotocin (STZ)-induced diabetic mice was investigated. RESEARCH DESIGN AND METHODS: The presence of the GLP-1R in lumbar dorsal root ganglion (DRG) was evaluated by immunohistochemical analyses. DRG neurons were dissected from C57BL6/J mice and cultured with or without Schwann cell-conditioned media in the presence or absence of GLP-1 (7-37) or exendin-4. Then neurite outgrowth was determined. In animal-model experiments, mice were made diabetic by STZ administration, and after 12 weeks of diabetes, exendin-4 (10 nmol/kg) was intraperitoneally administered once daily for 4 weeks. Peripheral nerve function was determined by the current perception threshold and motor and sensory nerve conduction velocity (MNCV and SNCV, respectively). Sciatic nerve blood flow (SNBF) and intraepidermal nerve fiber densities (IENFDs) also were evaluated. RESULTS: The expression of the GLP-1R in DRG neurons was confirmed. GLP-1 (7-37) and exendin-4 significantly promoted neurite outgrowth of DRG neurons. Both GLP-1R agonists accelerated the impaired neurite outgrowth of DRG neurons cultured with Schwann cell-conditioned media that mimicked the diabetic condition. At the doses used, exendin-4 had no effect on blood glucose or HbA(1c) levels. Hypoalgesia and delayed MNCV and SNCV in diabetic mice were improved by exendin-4 without affecting the reduced SNBF. The decreased IENFDs in sole skins of diabetic mice were ameliorated by exendin-4. CONCLUSIONS: Our findings indicate that exendin-4 ameliorates the severity of DPN, which may be achieved by its direct actions on DRG neurons and their axons.


Subject(s)
Diabetes Mellitus, Experimental/drug therapy , Diabetic Neuropathies/drug therapy , Hypoglycemic Agents/therapeutic use , Peptides/therapeutic use , Receptors, Glucagon/agonists , Venoms/therapeutic use , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/physiopathology , Diabetic Neuropathies/metabolism , Diabetic Neuropathies/physiopathology , Exenatide , Ganglia, Spinal/metabolism , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/pharmacology , Male , Mice , Neural Conduction/drug effects , Neural Conduction/physiology , Peptides/pharmacology , Schwann Cells/drug effects , Schwann Cells/metabolism , Sciatic Nerve/blood supply , Sciatic Nerve/drug effects , Sciatic Nerve/physiopathology , Venoms/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...