Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Biochem Biophys Res Commun ; 513(4): 974-982, 2019 06 11.
Article in English | MEDLINE | ID: mdl-31003779

ABSTRACT

AIMS/INTRODUCTION: Caloric restriction (CR) promotes longevity and exerts anti-aging effects by increasing Sirtuin production and activation. Gastric inhibitory polypeptide (GIP), a gastrointestinal peptide hormone, exerts various effects on pancreatic ß-cells and extra-pancreatic tissues. GIP promotes glucose-dependent augmentation of insulin secretion and uptake of nutrients into the adipose tissue. MATERIALS AND METHODS: Gipr-/- and Gipr+/+ mice were used for lifespan analysis, behavior experiments and gene expression of adipose tissue and muscles. 3T3-L1 differentiated adipocytes were used for Sirt1 and Nampt expression followed by treatment with GIP and α-lipoic acid. RESULTS: We observed that GIP receptor-knockout (Gipr-/-) mice fed normal diet showed an extended lifespan, increased exploratory and decreased anxiety-based behaviors, which are characteristic behavioral changes under CR. Moreover, Gipr-/- mice showed increased Sirt1 and Nampt expression in the adipose tissue. GIP suppressed α-lipoic acid-induced Sirt1 expression and activity in differentiated adipocytes. CONCLUSIONS: Although maintenance of CR is difficult, food intake and muscle endurance of Gipr-/- mice were similar to those of wild-type mice. Inhibition of GIP signaling may be a novel strategy to extend the lifespan of diabetic patients.


Subject(s)
Caloric Restriction , Gastric Inhibitory Polypeptide/antagonists & inhibitors , Longevity/physiology , Signal Transduction/physiology , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/metabolism , Animals , Cytokines/metabolism , Mice , Mice, Knockout , Nicotinamide Phosphoribosyltransferase/metabolism , Receptors, Gastrointestinal Hormone/genetics , Sirtuin 1/metabolism
2.
Kidney Int ; 90(4): 783-96, 2016 10.
Article in English | MEDLINE | ID: mdl-27475229

ABSTRACT

The role of stromal cell-derived factor-1 (SDF-1) in the pathogenesis of diabetic nephropathy and its modification by dipeptidyl peptidase-4 (DPP-4) inhibition are uncertain. Therefore, we studied this independent of glucagon-like peptide-1 receptor (GLP-1R) signaling using two Akita diabetic mouse models, the diabetic-resistant C57BL/6-Akita and diabetic-prone KK/Ta-Akita. Increased SDF-1 expression was found in glomerular podocytes and distal nephrons in the diabetic-prone mice, but not in kidneys from diabetic-resistant mice. The DPP-4 inhibitor linagliptin, but not the GLP-1R agonist liraglutide, further augmented renal SDF-1 expression in both Glp1r(+/+) and Glp1r(-/-) diabetic-prone mice. Along with upregulation of renal SDF-1 expression, the progression of albuminuria, glomerulosclerosis, periglomerular fibrosis, podocyte loss, and renal oxidative stress was suppressed in linagliptin-treated Glp1r(+/+) diabetic-prone mice. Linagliptin treatment increased urinary sodium excretion and attenuated the increase in glomerular filtration rate which reflects glomerular hypertension and hyperfiltration. In contrast, selective SDF-1 receptor blockade with AMD3100 reduced urinary sodium excretion and aggravated glomerular hypertension in the Glp1r(+/+) diabetic-prone mice. Thus, DPP-4 inhibition, independent of GLP-1R signaling, contributes to protection of the diabetic kidney through SDF-1-dependent antioxidative and antifibrotic effects and amelioration of adverse renal hemodynamics.


Subject(s)
Chemokine CXCL12/metabolism , Diabetes Mellitus, Type 1/complications , Diabetic Nephropathies/drug therapy , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Hypoglycemic Agents/therapeutic use , Kidney/pathology , Linagliptin/therapeutic use , Albuminuria/drug therapy , Albuminuria/urine , Animals , Benzylamines , Cyclams , Diabetes Mellitus, Type 1/genetics , Diabetic Nephropathies/urine , Disease Models, Animal , Female , Fibrosis , Glomerular Filtration Rate , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/genetics , Heterocyclic Compounds/pharmacology , Humans , Kidney/blood supply , Kidney/drug effects , Kidney/metabolism , Liraglutide/pharmacology , Male , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Receptors, CXCR4/antagonists & inhibitors , Up-Regulation
3.
Kidney Int ; 85(3): 579-89, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24152968

ABSTRACT

Glucagon-like peptide-1 (GLP-1) is a gut incretin hormone that has an antioxidative protective effect on various tissues. Here, we determined whether GLP-1 has a role in the pathogenesis of diabetic nephropathy using nephropathy-resistant C57BL/6-Akita and nephropathy-prone KK/Ta-Akita mice. By in situ hybridization, we found the GLP-1 receptor (GLP-1R) expressed in glomerular capillary and vascular walls, but not in tubuli, in the mouse kidney. Next, we generated C57BL/6-Akita Glp1r knockout mice. These mice exhibited higher urinary albumin levels and more advanced mesangial expansion than wild-type C57BL/6-Akita mice, despite comparable levels of hyperglycemia. Increased glomerular superoxide, upregulated renal NAD(P)H oxidase, and reduced renal cAMP and protein kinase A (PKA) activity were noted in the Glp1r knockout C57BL/6-Akita mice. Treatment with the GLP-1R agonist liraglutide suppressed the progression of nephropathy in KK/Ta-Akita mice, as demonstrated by reduced albuminuria and mesangial expansion, decreased levels of glomerular superoxide and renal NAD(P)H oxidase, and elevated renal cAMP and PKA activity. These effects were abolished by an adenylate cyclase inhibitor SQ22536 and a selective PKA inhibitor H-89. Thus, GLP-1 has a crucial role in protection against increased renal oxidative stress under chronic hyperglycemia, by inhibition of NAD(P)H oxidase, a major source of superoxide, and by cAMP-PKA pathway activation.


Subject(s)
Diabetic Nephropathies/etiology , Receptors, Glucagon/physiology , Signal Transduction/physiology , Animals , Cyclic AMP/analysis , Cyclic AMP-Dependent Protein Kinases/metabolism , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/metabolism , Glucagon-Like Peptide 1/analogs & derivatives , Glucagon-Like Peptide 1/therapeutic use , Glucagon-Like Peptide-1 Receptor , Kidney Glomerulus/immunology , Kidney Glomerulus/pathology , Liraglutide , Male , Mice, Inbred C57BL , NADPH Oxidases/antagonists & inhibitors , Nitric Oxide/analysis , Oxidative Stress , Receptors, Glucagon/agonists
4.
Nutrients ; 15(9)2023 Apr 22.
Article in English | MEDLINE | ID: mdl-37432141

ABSTRACT

The aim of this study was to investigate the effects of increased dietary protein in daily-life settings in Japan for 6 months on the activities of daily living (ADL) in adults aged 75 or older at nutritional risk. The study was an open-label, exploratory, randomized controlled trial conducted at seven hospitals in Japan. The study participants were adults aged 75 or older who were hospitalized for treatable cancer, pneumonia, fractures, and/or urinary-tract infection at nutritional risk. The primary outcome was change in grip strength, skeletal muscle, and ADL indices (Barthel index, Lawton score). One hundred sixty-nine patients were randomly assigned to the intensive care (IC) or standard care (SC) group; the protein intake goals (g/kgw/day) were 1.5 for IC and 1.0 for SC. There was a significant improvement in grip strength only in the IC group (1.1 kg: 95% CI 0.1 to 2.1) (p = 0.02). While the skeletal muscle index and ADL indices were not significantly improved in either group, the improvement ratio tended to be greater in the IC group. There was no decrease in renal function in either group. Thus, intervention of increased dietary protein in daily-life settings for 6 months in adults aged 75 or older with treatable cancer, pneumonia, fractures, and/or urinary-tract infection and at nutritional risk may be effective in ameliorating loss of muscle strength.


Subject(s)
Activities of Daily Living , Fractures, Bone , Humans , Adult , Dietary Proteins , Research Design , Critical Care
5.
Biochem Biophys Res Commun ; 404(1): 115-20, 2011 Jan 07.
Article in English | MEDLINE | ID: mdl-21095180

ABSTRACT

Gastric inhibitory polypeptide (GIP) is released from the small intestine upon meal ingestion and increases insulin secretion from pancreatic ß cells. Although the GIP receptor is known to be expressed in small intestine, the effects of GIP in small intestine are not fully understood. This study was designed to clarify the effect of GIP on intestinal glucose absorption and intestinal motility. Intestinal glucose absorption in vivo was measured by single-pass perfusion method. Incorporation of [(14)C]-glucose into everted jejunal rings in vitro was used to evaluate the effect of GIP on sodium-glucose co-transporter (SGLT). Motility of small intestine was measured by intestinal transit after oral administration of a non-absorbed marker. Intraperitoneal administration of GIP inhibited glucose absorption in wild-type mice in a concentration-dependent manner, showing maximum decrease at the dosage of 50 nmol/kg body weight. In glucagon-like-peptide-1 (GLP-1) receptor-deficient mice, GIP inhibited glucose absorption as in wild-type mice. In vitro examination of [(14)C]-glucose uptake revealed that 100 nM GIP did not change SGLT-dependent glucose uptake in wild-type mice. After intraperitoneal administration of GIP (50 nmol/kg body weight), small intestinal transit was inhibited to 40% in both wild-type and GLP-1 receptor-deficient mice. Furthermore, a somatostatin receptor antagonist, cyclosomatostatin, reduced the inhibitory effect of GIP on both intestinal transit and glucose absorption in wild-type mice. These results demonstrate that exogenous GIP inhibits intestinal glucose absorption by reducing intestinal motility through a somatostatin-mediated pathway rather than through a GLP-1-mediated pathway.


Subject(s)
Gastric Inhibitory Polypeptide/pharmacology , Gastrointestinal Motility/drug effects , Glucose/metabolism , Intestinal Absorption/drug effects , Jejunum/drug effects , Animals , Glucagon-Like Peptide-1 Receptor , Jejunum/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Perfusion , Receptors, Glucagon/genetics , Somatostatin/pharmacology
6.
Nat Med ; 8(7): 738-42, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12068290

ABSTRACT

Secretion of gastric inhibitory polypeptide (GIP), a duodenal hormone, is primarily induced by absorption of ingested fat. Here we describe a novel pathway of obesity promotion via GIP. Wild-type mice fed a high-fat diet exhibited both hypersecretion of GIP and extreme visceral and subcutaneous fat deposition with insulin resistance. In contrast, mice lacking the GIP receptor (Gipr(-/-)) fed a high-fat diet were clearly protected from both the obesity and the insulin resistance. Moreover, double-homozygous mice (Gipr(-/-), Lep(ob)/Lep(ob)) generated by crossbreeding Gipr(-/-) and obese ob/ob (Lep(ob)/Lep(ob)) mice gained less weight and had lower adiposity than Lep(ob)/Lep(ob) mice. The Gipr(-/-) mice had a lower respiratory quotient and used fat as the preferred energy substrate, and were thus resistant to obesity. Therefore, GIP directly links overnutrition to obesity and it is a potential target for anti-obesity drugs.


Subject(s)
Adipose Tissue/physiology , Gastric Inhibitory Polypeptide/physiology , Obesity/prevention & control , Receptors, Gastrointestinal Hormone/physiology , Signal Transduction/physiology , Adipose Tissue/anatomy & histology , Animals , Body Weight , Crosses, Genetic , Dietary Fats , Gastric Inhibitory Polypeptide/deficiency , Gastric Inhibitory Polypeptide/genetics , Mice , Mice, Knockout , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Gastrointestinal Hormone/genetics
7.
J Clin Invest ; 117(1): 143-52, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17187081

ABSTRACT

The incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) control glucose homeostasis through well-defined actions on the islet beta cell via stimulation of insulin secretion and preservation and expansion of beta cell mass. We examined the importance of endogenous incretin receptors for control of glucose homeostasis through analysis of Glp1r(-/-), Gipr(-/-), and double incretin receptor knockout (DIRKO) mice fed a high-fat (HF) diet. DIRKO mice failed to upregulate levels of plasma insulin, pancreatic insulin mRNA transcripts, and insulin content following several months of HF feeding. Both single incretin receptor knockout and DIRKO mice exhibited resistance to diet-induced obesity, preservation of insulin sensitivity, and increased energy expenditure associated with increased locomotor activity. Moreover, plasma levels of plasminogen activator inhibitor-1 and resistin failed to increase significantly in DIRKO mice after HF feeding, and the GIP receptor agonist [D-Ala(2)]GIP, but not the GLP-1 receptor agonist exendin-4, increased the levels of plasma resistin in studies of both acute and chronic administration. These findings extend our understanding of how endogenous incretin circuits regulate glucose homeostasis independent of the beta cell via control of adipokine secretion and energy expenditure.


Subject(s)
Body Weight/physiology , Energy Metabolism/physiology , Receptors, Glucagon/physiology , Animals , Glucagon-Like Peptide-1 Receptor , Homeostasis , Insulin/genetics , Islets of Langerhans/physiology , Mice , Mice, Knockout , RNA, Messenger/genetics , Receptors, Glucagon/deficiency , Receptors, Glucagon/genetics , Transcription, Genetic
8.
Endocrinology ; 149(2): 574-9, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18039776

ABSTRACT

Gastrointestinal hormones including gastric inhibitory polypeptide (GIP), glucagon-like peptide (GLP)-1, and GLP-2 are secreted immediately after meal ingestion, and GIP and GLP-2 have been shown to regulate bone turnover. We hypothesize that endogenous GLP-1 may also be important for control of skeletal homeostasis. We investigated the role of GLP-1 in the regulation of bone metabolism using GLP-1 receptor knockout (Glp-1r(-/-)) mice. A combination of bone density and histomorphometry, osteoclast activation studies, biochemical analysis of calcium and PTH, and RNA analysis was used to characterize bone and mineral homeostasis in Glp-1r(-/-) and Glp-1r(+/+) littermate controls. Glp-1r(-/-) mice have cortical osteopenia and bone fragility by bone densitometry as well as increased osteoclastic numbers and bone resorption activity by bone histomorphometry. Although GLP-1 had no direct effect on osteoclasts and osteoblasts, Glp-1r(-/-) mice exhibited higher levels of urinary deoxypyridinoline, a marker of bone resorption, and reduced levels of calcitonin mRNA transcripts in the thyroid. Moreover, calcitonin treatment effectively suppressed urinary levels of deoxypyridinoline in Glp-1r(-/-), mice and the GLP-1 receptor agonist exendin-4 increased calcitonin gene expression in the thyroid of wild-type mice. These findings establish an essential role for endogenous GLP-1 receptor signaling in the control of bone resorption, likely through a calcitonin-dependent pathway.


Subject(s)
Bone Resorption/metabolism , Bone Resorption/physiopathology , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Absorptiometry, Photon , Animals , Bone Resorption/diagnostic imaging , Calcitonin/metabolism , Calcium/metabolism , Exenatide , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/cytology , Osteoblasts/physiology , Osteoclasts/cytology , Osteoclasts/physiology , Peptides/metabolism , Peptides/pharmacology , Signal Transduction/physiology , Tibia/diagnostic imaging , Tibia/physiology , Venoms/metabolism , Venoms/pharmacology
9.
Biochem Biophys Res Commun ; 364(1): 175-80, 2007 Dec 07.
Article in English | MEDLINE | ID: mdl-17937928

ABSTRACT

Aging is associated with increased fat mass and decreased lean mass, which is strongly associated with the development of insulin resistance. Gastric inhibitory polypeptide (GIP) is known to promote efficient storage of ingested nutrients into adipose tissue; we examined aging-associated changes in body composition using 10-week-old and 50-week-old wild-type (WT) and GIP receptor knockout (Gipr-/-) mice on a normal diet, which show no difference in body weight. We found that Gipr-/- mice showed significantly reduced fat mass without reduction of lean mass or food intake, while WT mice showed increased fat mass and decreased lean mass associated with aging. Moreover, aged Gipr-/- mice showed improved insulin sensitivity, which is associated with amelioration in glucose tolerance, higher plasma adiponectin levels, and increased spontaneous physical activity. We therefore conclude that genetic inactivation of GIP signaling can prevent the development of aging-associated insulin resistance through body composition changes.


Subject(s)
Aging/physiology , Body Composition/physiology , Gastric Inhibitory Polypeptide/genetics , Gastric Inhibitory Polypeptide/physiology , Insulin Resistance/physiology , Signal Transduction/physiology , Adipokines/blood , Adipose Tissue/anatomy & histology , Animals , Behavior, Animal , Blood Glucose/metabolism , Glucose Tolerance Test , Male , Mice , Mice, Knockout , Motor Activity
10.
Mol Endocrinol ; 20(7): 1644-51, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16469773

ABSTRACT

Calcium plays a fundamental role as second messenger in intracellular signaling and bone serves as the body's calcium reserve to tightly maintain blood calcium levels. Calcium in ingested meal is the main supply and inadequate calcium intake causes osteoporosis and bone fracture. Here, we describe a novel mechanism of how ingested calcium is deposited on bone. Meal ingestion elicits secretion of the gut hormone gastric inhibitory polypeptide (GIP) from endocrine K cells in the duodenum. Bone histomorphometrical analyses revealed that bone formation parameters in the mice lacking GIP receptor (GIPR(-/-)) were significantly lower than those of wild-type (GIPR(+/+)) mice, and that the number of osteoclasts, especially multinuclear osteoclasts, was significantly increased in GIPR(-/-) mice, indicating that GIPR(-/-) mice have high-turnover osteoporosis. In vitro examination showed the percentage of osteoblastic cells undergoing apoptosis to be significantly decreased in the presence of GIP. Because GIPR(-/-) mice exhibited an increased plasma calcium concentration after meal ingestion, GIP directly links calcium contained in meal to calcium deposition on bone.


Subject(s)
Eating/physiology , Gastric Inhibitory Polypeptide/physiology , Osteogenesis/physiology , Animals , Apoptosis Regulatory Proteins/metabolism , Bone Resorption/congenital , Calcium/blood , Calcium/metabolism , Gastric Inhibitory Polypeptide/deficiency , Gastric Inhibitory Polypeptide/metabolism , Male , Mice , Mice, Transgenic , Models, Biological , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/pathology , Signal Transduction , Tibia/anatomy & histology , Tibia/growth & development
11.
Endocrinology ; 158(7): 2134-2144, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28430907

ABSTRACT

In addition to overeating, starvation also reduces fecundity in mammals. However, little is known about the molecular mechanisms linking food intake to fertility, especially in males. Gastric inhibitory polypeptide (GIP), which is released from intestinal K-cells after meal ingestion, stimulates insulin secretion from pancreatic ß-cells through the action of incretin and has several extrapancreatic effects. Here, we identified GIP receptor (Gipr) expression in mouse spermatids. Microarray analysis revealed that pregnancy-specific glycoprotein 17 (Psg17), a potential CD9-binding partner, was significantly decreased in GIP receptor-knockout (Gipr-/-) testes. Glycosylphosphatidylinositol-anchored PSG17 was expressed on the surface of acrosome-reacted sperm, and Gipr-/- sperm led to a lower fertilization rate in vitro, compared with that of Gipr+/+ sperm, both in the absence and presence of the zona pellucida. Plasma GIP concentrations and Psg17 messenger RNA (mRNA) were immediately increased in the testis after a single meal, whereas ingestion of a chronic high-fat diet markedly decreased Gipr and Psg17 mRNA. These results suggest that reduced GIP signaling, by decreased GIP levels or the downregulation of Gipr, is associated with the reduction of fecundity due to starvation or overeating. Thus, proper regulation of GIP signaling in the testis could be a potential unique therapeutic target for male infertility in obese and diabetic individuals.


Subject(s)
Eating/physiology , Gastric Inhibitory Polypeptide/physiology , Glycoproteins/physiology , Pregnancy Proteins/physiology , Receptors, Gastrointestinal Hormone/physiology , Sperm-Ovum Interactions/genetics , Animals , Female , Fertility/genetics , Gastric Inhibitory Polypeptide/genetics , Glycoproteins/genetics , Intercellular Signaling Peptides and Proteins , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Pregnancy Proteins/genetics , Receptors, Gastrointestinal Hormone/genetics , Signal Transduction/genetics , Testis/metabolism
12.
Lancet ; 365(9471): 1642-4, 2005.
Article in English | MEDLINE | ID: mdl-15889479

ABSTRACT

Rising demand for islet transplantation will lead to severe donor shortage in the near future, especially in countries where cadaveric organ donation is scarce. We undertook a successful transplantation of living-donor islets for unstable diabetes. The recipient was a 27-year-old woman who had had brittle, insulin-dependent diabetes mellitus for 12 years. The donor, who was a healthy 56-year-old woman and mother of the recipient, underwent a distal pancreatectomy. After isolation, 408 114 islet equivalents were transplanted immediately. The transplants functioned immediately and the recipient became insulin-independent 22 days after the operation. The donor had no complications and both women showed healthy glucose tolerance. Transplantation of living-donor islets from the distal pancreas can be sufficient to reverse brittle diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Living Donors , Pancreatectomy/methods , Tissue and Organ Harvesting/methods , Adult , Diabetes Mellitus, Type 1/complications , Female , Humans , Middle Aged , Pancreatitis/complications
13.
Ann N Y Acad Sci ; 1079: 335-9, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17130575

ABSTRACT

We performed the world's first successful living donor islet transplantation for unstable diabetes. A total of 408,114 islet equivalents were isolated from half a living pancreas and transplanted immediately to the recipient who was a 27-year-old female. The donor was a 56-year-old female in good health, mother of the recipient. The islets functioned immediately, and the recipient was weaned completely from insulin on the 22nd posttransplant day, and has maintained excellent glycemic control since. The donor was discharged on the 18th postoperative day with normal oral glucose tolerance test and without complications. Living donor islet transplantation could cure one insulin-dependent diabetes mellitus patients with a single donor. There are some advantages in the living donor islet transplantation: (a) living donor can alleviate the issue of donor shortage; (b) highly potent islets can be isolated from a living donor; and (c) the recipient can be treated with immunosuppressant and controlled blood glucose level tightly prior to the transplantation. These are important factors in overcoming the obstacles limiting islet transplantation. We believe that the living donor islet transplantation may become an additional option in treating insulin-dependent diabetes.


Subject(s)
Diabetes Mellitus, Type 1/surgery , Islets of Langerhans Transplantation , Living Donors , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/therapeutic use , Basiliximab , Blood Glucose/metabolism , C-Peptide/blood , Diabetes Mellitus, Type 1/drug therapy , Female , Follow-Up Studies , Glucose Tolerance Test , Glycated Hemoglobin/analysis , Humans , Hypoglycemic Agents/therapeutic use , Infliximab , Insulin/therapeutic use , Middle Aged , Postoperative Period , Preoperative Care , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/therapeutic use , Sirolimus/administration & dosage , Sirolimus/therapeutic use , Tacrolimus/administration & dosage , Tacrolimus/therapeutic use , Time Factors , Treatment Outcome
14.
Diabetes Res Clin Pract ; 74(3): 222-6, 2006 Dec.
Article in English | MEDLINE | ID: mdl-16707190

ABSTRACT

Evaluation of a patient's pancreatic beta-cell function is important in both diagnosis and treatment of diabetes. We sought to determine beta-cell function with a single sampling of blood. Examination of fasting blood glucose (F-BG, mM) and C-peptide (F-CPR, nM) levels in seven post-islet-transplanted states of four patients revealed a linear relationship between F-BG and F-CPR. Assuming that normal subjects aged <40 years have 100% pancreatic beta-cell function, we developed the secretory units of islets in transplantation (SUIT) as an index of beta-cell function by the formula: 250 x F-CPR/(F-BG-3.43). The SUIT index was correlated with the stimulated C-peptide levels not only in islet-transplanted patients (R2 = 0.68, P < 0.05) but also in type 2 patients (R2 = 0.34, P < 0.001). Since the SUIT index can be calculated from data obtained at a single fasting blood sampling and predict the pancreatic beta-cell function, the formula may be a useful tool in clinical management of diabetes.


Subject(s)
Diabetes Mellitus, Type 2/therapy , Islets of Langerhans Transplantation , Islets of Langerhans/metabolism , Blood Glucose , Humans , Insulin/therapeutic use
15.
J Diabetes Investig ; 7 Suppl 1: 76-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27186360

ABSTRACT

The hormonal factors implicated as transmitters of signals from the gut to pancreatic ß-cells are referred to as incretins. Gastric inhibitory polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) are incretins. In addition to the insulinotropic effects, we have shown, using the GIP receptor and GLP-1 receptor-deficient mice, that GIP and GLP-1 have direct actions on adipocytes and the kidney, respectively. Because GIP receptors and GLP-1 receptors are differentially expressed in a tissue-specific manner, GIP and GLP-1 have specific physiological activities, and further comprehensive characterization of the extrapancreatic actions of GIP and GLP-1 is anticipated, as dipeptidyl peptidase IV inhibitors activate both GIP and GLP-1 signaling.


Subject(s)
Enteroendocrine Cells/metabolism , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Incretins/metabolism , Animals , Glucagon-Like Peptide-1 Receptor/metabolism , Glucose/metabolism , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice , Satiation/physiology
16.
J Diabetes Investig ; 7(4): 497-505, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27181102

ABSTRACT

AIMS/INTRODUCTION: The involvement of glucose-dependent insulinotropic polypeptide (GIP) on inflammation was explored in atherosclerosis and adipose tissue. Periodontal disease is a chronic inflammatory disease, and is considered one of the diabetic complications. In the present study, to examine the effect of GIP on periodontitis, we induced experimental periodontitis in glucose-dependent insulinotropic polypeptide receptor-knockout mice (GIPRKO). We also investigated the anti-inflammatory effect of GIP in a culture system. MATERIALS AND METHODS: Experimental periodontitis was induced by ligature wire in GIPRKO and C57BL/C mice. Two weeks after the ligature, immunohistological evaluation and inflammatory messenger ribonucleic acid expression in the gingiva was examined. To elucidate the role of GIP in inflammation, the effects of GIP on lipopolysaccharide-induced gene expressions in THP-1 cells were evaluated. RESULTS: Periodontitis increased inflammatory cell infiltration, macrophage accumulation and tumor necrosis factor-α and nitric oxide synthase gene expressions in the gingiva. Periodontitis in GIPRKO showed a marked increase of inflammatory cells in the gingivomucosal tissue. Mac-1-positive macrophages and the inflammatory gene expressions were significantly increased in periodontitis in GIPRKO compared with C57BL/C mice periodontitis. Immunohistochemical staining confirmed that GIP receptors were expressed in residual and infiltrated Mac-1-positive macrophages. The in vitro study showed that GIP suppressed lipopolysaccharide-induced tumor necrosis factor-α and nitric oxide synthase gene expression in a dose-dependent manner. Furthermore, the inhibitory effect of GIP on lipopolysaccharide-induced inflammatory gene expressions was at least partially through cyclic adenosine monophosphate/protein kinase A pathway. CONCLUSIONS: These results suggest the beneficial effects of GIP on periodontal disease. In diabetic patients, GIP is expected to have a direct anti-inflammatory effect on periodontitis in addition to its glucose-lowering effect.


Subject(s)
Gastric Inhibitory Polypeptide/physiology , Periodontitis/physiopathology , Receptors, Gastrointestinal Hormone/physiology , Animals , Cell Culture Techniques , Cytokines/metabolism , Disease Models, Animal , Gastric Inhibitory Polypeptide/metabolism , Humans , Inflammation Mediators/metabolism , Macrophages/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase Type II/metabolism , Periodontitis/metabolism , Receptors, Gastrointestinal Hormone/genetics , Tumor Necrosis Factor-alpha/metabolism
17.
Diabetes ; 53(5): 1326-35, 2004 May.
Article in English | MEDLINE | ID: mdl-15111503

ABSTRACT

Glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide 1 (GLP-1) are gut-derived incretins that potentiate glucose clearance following nutrient ingestion. Elimination of incretin receptor action in GIPR(-/-) or GLP-1R(-/-) mice produces only modest impairment in glucose homeostasis, perhaps due to compensatory upregulation of the remaining incretin. We have now studied glucose homeostasis in double incretin receptor knockout (DIRKO) mice. DIRKO mice exhibit normal body weight and fail to exhibit an improved glycemic response after exogenous administration of GIP or the GLP-1R agonist exendin-4. Plasma glucagon and the hypoglycemic response to exogenous insulin were normal in DIRKO mice. Glycemic excursion was abnormally increased and levels of glucose-stimulated insulin secretion were decreased following oral but not intraperitoneal glucose challenge in DIRKO compared with GIPR(-/-) or GLP-1R(-/-) mice. Similarly, glucose-stimulated insulin secretion and the response to forskolin were well preserved in perifused DIRKO islets. Although the dipeptidyl peptidase-IV (DPP-IV) inhibitors valine pyrrolidide (Val-Pyr) and SYR106124 lowered glucose and increased plasma insulin in wild-type and single incretin receptor knockout mice, the glucose-lowering actions of DPP-IV inhibitors were eliminated in DIRKO mice. These findings demonstrate that glucose-stimulated insulin secretion is maintained despite complete absence of both incretin receptors, and they delineate a critical role for incretin receptors as essential downstream targets for the acute glucoregulatory actions of DPP-IV inhibitors.


Subject(s)
Dipeptidyl Peptidase 4/drug effects , Enzyme Inhibitors/pharmacology , Glucose/metabolism , Intestines/physiology , Islets of Langerhans/physiology , Receptors, Gastrointestinal Hormone/physiology , Receptors, Glucagon/physiology , Animals , Exenatide , Glucagon-Like Peptide-1 Receptor , Homeostasis , Insulin/metabolism , Insulin Secretion , Mice , Mice, Inbred C57BL , Mice, Knockout , Organic Chemicals/pharmacology , Peptides/pharmacology , Pyrroles/pharmacology , Receptors, Gastrointestinal Hormone/agonists , Receptors, Gastrointestinal Hormone/deficiency , Receptors, Glucagon/agonists , Receptors, Glucagon/deficiency , Valine/pharmacology , Venoms/pharmacology
18.
Eur J Endocrinol ; 151(3): 407-12, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15362972

ABSTRACT

OBJECTIVE: ATP-sensitive K(+) (K(ATP)) channels in pancreatic beta-cells are crucial in the regulation of glucose-induced insulin secretion. Recently, K(ATP) channel-deficient mice were generated by genetic disruption of Kir6.2, the pore-forming component of K(ATP) channels, but the mice still showed a significant insulin response after oral glucose loading in vivo. Gastric inhibitory polypeptide (GIP) is a physiological incretin that stimulates insulin release upon ingestion of nutrients. To determine if GIP is the insulinotropic factor in insulin secretion in K(ATP) channel-deficient mice, we generated double-knockout Kir6.2 and GIP receptor null mice and compared them with Kir6.2 knockout mice. METHODS: Double-knockout mice were generated by intercrossing Kir6.2-knockout mice with GIP receptor-knockout mice. An oral glucose tolerance test, insulin tolerance test and batch incubation study of pancreatic islets were performed on double-knockout mice and Kir6.2-knockout mice. RESULTS: Fasting glucose and insulin levels were similar in both groups. After oral glucose loading, blood glucose levels of double-knockout mice became elevated compared with Kir6.2-knockout mice, especially at 15 min (345+/-10 mg/dl vs 294+/-20 mg/dl, P<0.05) and 30 min (453+/-20 mg/dl vs 381+/-26 mg/dl, P<0.05). The insulin response was almost completely lost in double-knockout mice, although insulin secretion from isolated islets was stimulated by another incretin, glucagon-like peptide-1 in the double-knockout mice. Double-knockout mice and Kir6.2-knockout mice were similarly insulin sensitive as assessed by the insulin tolerance test. CONCLUSION: GIP is the major insulinotropic factor in the secretion of insulin in response to glucose load in K(ATP) channel-deficient mice.


Subject(s)
Gastric Inhibitory Polypeptide/metabolism , Hypoglycemic Agents/metabolism , Insulin/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Receptors, Gastrointestinal Hormone/genetics , Animals , Blood Glucose , Glucose/pharmacokinetics , Glucose Intolerance/metabolism , Glucose Intolerance/physiopathology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Insulin Secretion , Islets of Langerhans/metabolism , Jejunum/metabolism , Mice , Mice, Knockout
19.
Nihon Rinsho ; 62(6): 1077-84, 2004 Jun.
Article in Japanese | MEDLINE | ID: mdl-15206144

ABSTRACT

Gastric inhibitory polypeptide(GIP) is a gastrointestinal peptide hormone, which is secreted from duodenal endocrine K cells after absorption of glucose or fat. It is well known as an incretin. To determine the further role of GIP in vivo, we generated GIP receptor-knockout mice. The mice showed higher blood glucose levels with impaired initial insulin response after oral glucose load. Even after high-fat diet, knockout mice lack compensatory insulin secretion, and showed no hyper-insulinemia. Moreover, knockout mice fed a high-fat diet were clearly protected from both the obesity and the insulin resistance. Therefore, GIP directly links glucose tolerance and over-nutrition to obesity and it is a potential target for the treatment for the metabolic syndrome.


Subject(s)
Gastric Inhibitory Polypeptide/deficiency , Metabolic Syndrome/metabolism , Peptide Fragments/deficiency , Animals , Disease Models, Animal , Mice , Mice, Knockout
20.
Diabetes Res Clin Pract ; 106(3): 538-47, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25451890

ABSTRACT

AIM: To assess changes in circulating incretin levels and body fat compositions with initial combination therapy with α-glucosidase inhibitor and dipeptidyl peptidase-4 inhibitor in patients with type 2 diabetes (T2D). METHODS: In this multicenter open-label 24-week trial, Japanese over-weight (BMI ≥ 25 kg/m(2)) patients with T2D not taking medication or taking metformin and/or sulfonylurea were randomly assigned to receive either 50mg of miglitol three times a day (M, n=14), 50mg of sitagliptin once a day (S, n=14), or a combination of both (M+S, n=13). Changes in plasma incretin levels during a meal tolerance test (MTT) and body fat composition with impedance method were evaluated. RESULTS: During MTT, postprandial plasma glucose levels decreased more after M+S than after M or S, and postprandial serum insulin levels decreased significantly after M and M+S whereas they increased after S. After M, active gastric inhibitory polypeptide (aGIP) decreased significantly at 30 min despite a significant increase at 120 min. After S, aGIP levels increased significantly throughout the MTT. After M+S, aGIP increased significantly at 0 and 120 min despite of significant decrease at 30 min. M+S further enhanced postprandial active glucagon-like peptide-1 levels during MTT than S did. Total body fat mass decreased significantly after M and M+S. Visceral fat mass decreased significantly only after M+S. Serum adiponectin increased significantly only after M+S. CONCLUSIONS: In over-weight patients with T2D, M+S may have a beneficial effect on adiposity with relation to these different effects on two incretins.


Subject(s)
1-Deoxynojirimycin/analogs & derivatives , Diabetes Mellitus, Type 2/therapy , Incretins/blood , Intra-Abdominal Fat/metabolism , Overweight/complications , Pyrazines/administration & dosage , Triazoles/administration & dosage , 1-Deoxynojirimycin/administration & dosage , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Hypoglycemic Agents/administration & dosage , Intra-Abdominal Fat/drug effects , Japan/epidemiology , Male , Middle Aged , Overweight/epidemiology , Overweight/metabolism , Postprandial Period , Sitagliptin Phosphate , Time Factors , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL