Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Diabetes ; 73(3): 474-489, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38064504

ABSTRACT

Genome-wide association studies have identified several gene polymorphisms, including UBE2E2, associated with type 2 diabetes. Although UBE2E2 is one of the ubiquitin-conjugating enzymes involved in the process of ubiquitin modifications, the pathophysiological roles of UBE2E2 in metabolic dysfunction are not yet understood. Here, we showed upregulated UBE2E2 expression in the islets of a mouse model of diet-induced obesity. The diabetes risk allele of UBE2E2 (rs13094957) in noncoding regions was associated with upregulation of UBE2E2 mRNA in the human pancreas. Although glucose-stimulated insulin secretion was intact in the isolated islets, pancreatic ß-cell-specific UBE2E2-transgenic (TG) mice exhibited reduced insulin secretion and decreased ß-cell mass. In TG mice, suppressed proliferation of ß-cells before the weaning period and while receiving a high-fat diet was accompanied by elevated gene expression levels of p21, resulting in decreased postnatal ß-cell mass expansion and compensatory ß-cell hyperplasia, respectively. In TG islets, proteomic analysis identified enhanced formation of various types of polyubiquitin chains, accompanied by increased expression of Nedd4 E3 ubiquitin protein ligase. Ubiquitination assays showed that UBE2E2 mediated the elongation of ubiquitin chains by Nedd4. The data suggest that UBE2E2-mediated ubiquitin modifications in ß-cells play an important role in regulating glucose homeostasis and ß-cell mass.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Insulin-Secreting Cells , Mice , Animals , Humans , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Genome-Wide Association Study , Proteomics , Insulin-Secreting Cells/metabolism , Glucose/metabolism , Mice, Transgenic , Diet, High-Fat/adverse effects , Ubiquitins/genetics , Ubiquitins/metabolism , Insulin/metabolism
2.
Nature ; 621(7978): 389-395, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648852

ABSTRACT

Insulin resistance is the primary pathophysiology underlying metabolic syndrome and type 2 diabetes1,2. Previous metagenomic studies have described the characteristics of gut microbiota and their roles in metabolizing major nutrients in insulin resistance3-9. In particular, carbohydrate metabolism of commensals has been proposed to contribute up to 10% of the host's overall energy extraction10, thereby playing a role in the pathogenesis of obesity and prediabetes3,4,6. Nevertheless, the underlying mechanism remains unclear. Here we investigate this relationship using a comprehensive multi-omics strategy in humans. We combine unbiased faecal metabolomics with metagenomics, host metabolomics and transcriptomics data to profile the involvement of the microbiome in insulin resistance. These data reveal that faecal carbohydrates, particularly host-accessible monosaccharides, are increased in individuals with insulin resistance and are associated with microbial carbohydrate metabolisms and host inflammatory cytokines. We identify gut bacteria associated with insulin resistance and insulin sensitivity that show a distinct pattern of carbohydrate metabolism, and demonstrate that insulin-sensitivity-associated bacteria ameliorate host phenotypes of insulin resistance in a mouse model. Our study, which provides a comprehensive view of the host-microorganism relationships in insulin resistance, reveals the impact of carbohydrate metabolism by microbiota, suggesting a potential therapeutic target for ameliorating insulin resistance.


Subject(s)
Carbohydrate Metabolism , Gastrointestinal Microbiome , Insulin Resistance , Animals , Humans , Mice , Diabetes Mellitus, Type 2/metabolism , Gastrointestinal Microbiome/physiology , Insulin Resistance/physiology , Monosaccharides/metabolism , Insulin/metabolism , Metabolic Syndrome/metabolism , Feces/chemistry , Feces/microbiology , Metabolomics
3.
J Am Heart Assoc ; 10(23): e020760, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34796738

ABSTRACT

Background It is uncertain whether risk classification under the nationwide program on screening and lifestyle modification for metabolic syndrome captures well high-risk individuals who could benefit from lifestyle interventions. We examined the validity of risk classification by linking the incidence of cardiovascular disease (CVD). Methods and Results Individual-level data of 29 288 Japanese individuals aged 40 to 74 years without a history of CVD from 10 prospective cohort studies were used. Metabolic syndrome was defined as the presence of high abdominal obesity and/or overweight plus risk factors such as high blood pressure, high triglyceride or low high-density lipoprotein cholesterol levels, and high blood glucose levels. The risk categories for lifestyle intervention were information supply only, motivation-support intervention, and intensive support intervention. Sex- and age-specific hazard ratios and population attributable fractions of CVD, which were also further adjusted to consider non-high density lipoprotein cholesterol levels, were estimated with reference to nonobese/overweight individuals, using Cox proportional hazard regression. Since the reference category included those with risk factors, we set a supernormal group (nonobese/overweight with no risk factor) as another reference. We documented 1023 incident CVD cases (565 men and 458 women). The adjusted CVD risk was 60% to 70% higher in men and women aged 40 to 64 years receiving an intensive support intervention, and 30% higher in women aged 65 to 74 years receiving a motivation-support intervention, compared with nonobese/overweight individuals. The population attributable fractions in men and women aged 40 to 64 years receiving an intensive support intervention were 17.7% and 6.6%, respectively, while that in women aged 65 to 74 years receiving a motivation-support intervention was 9.4%. Compared with the supernormal group, nonobese/overweight individuals with risk factors had similar hazard ratios and population attributable fractions as individuals with metabolic syndrome. Conclusions Similar CVD excess and attributable risks among individuals with metabolic syndrome components in the absence and presence of obesity/overweight imply the need for lifestyle modification in both high-risk groups.


Subject(s)
Cardiovascular Diseases , Metabolic Syndrome , Obesity , Adult , Aged , Cardiovascular Diseases/epidemiology , Female , Humans , Incidence , Japan/epidemiology , Male , Metabolic Syndrome/epidemiology , Middle Aged , Obesity/epidemiology , Prevalence , Prospective Studies , Risk Assessment
4.
Diabetes ; 70(8): 1640-1653, 2021 08.
Article in English | MEDLINE | ID: mdl-33980693

ABSTRACT

Insulin receptor substrate-1 (Irs1) is one of the major substrates for insulin receptor and insulin-like growth factor-1 (IGF-1) receptor tyrosine kinases. Systemic Irs1-deficient mice show growth retardation, with resistance to insulin and IGF-1, although the underlying mechanisms remain poorly understood. For this study, we generated mice with brain-specific deletion of Irs1 (NIrs1KO mice). The NIrs1KO mice exhibited lower body weights, shorter bodies and bone lengths, and decreased bone density. Moreover, the NIrs1KO mice exhibited increased insulin sensitivity and glucose utilization in the skeletal muscle. Although the ability of the pituitary to secrete growth hormone (GH) remained intact, the amount of hypothalamic growth hormone-releasing hormone (GHRH) was significantly decreased and, accordingly, the pituitary GH mRNA expression levels were impaired in these mice. Plasma GH and IGF-1 levels were also lower in the NIrs1KO mice. The expression levels of GHRH protein in the median eminence, where Irs1 antibody staining is observed, were markedly decreased in the NIrs1KO mice. In vitro, neurite elongation after IGF-1 stimulation was significantly impaired by Irs1 downregulation in the cultured N-38 hypothalamic neurons. In conclusion, brain Irs1 plays important roles in the regulation of neurite outgrowth of GHRH neurons, somatic growth, and glucose homeostasis.


Subject(s)
Brain/metabolism , Growth Disorders/genetics , Growth Hormone-Releasing Hormone/genetics , Hypothalamus/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance/physiology , Adipose Tissue, White/metabolism , Animals , Glucose/metabolism , Growth Disorders/metabolism , Growth Hormone/blood , Growth Hormone-Releasing Hormone/metabolism , Homeostasis/physiology , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Neurons/metabolism
5.
J Diabetes Investig ; 12(2): 266-276, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32621777

ABSTRACT

AIMS/INTRODUCTION: To prevent diabetic complications, strict glucose control and frequent monitoring of blood glucose levels with invasive methods are necessary. We considered the monitoring of tear glucose levels might be a possible method for non-invasive glucose monitoring. To develop tear glucose monitoring for clinical application, we investigated the precise correlation between the blood and tear glucose concentrations. MATERIALS AND METHODS: A total of 10 participants and 20 participants with diabetes were admitted, and blood and tear samples were collected. Before statistical analysis, we eliminated tear samples contaminated with blood. We observed the daily blood and tear glucose dynamics, and carried out a random intercept model analysis to examine the association between the blood and tear glucose concentrations. RESULTS: Tear occult blood tests showed that the tear glucose concentrations and their variation increased in both participants with and without diabetes as contamination of blood increased. In both participants with and without diabetes, fluctuations of the plasma glucose concentrations were observed depending on the timing of collection of the samples, and the dynamics of the tear glucose concentrations paralleled those of the plasma glucose concentrations. The random intercept model analysis showed a significant association between the plasma and tear glucose concentrations in participants with diabetes (P < 0.001). This association still existed even after adjusting for the glycated hemoglobin levels and the prandial state (P < 0.001). CONCLUSIONS: It is important to eliminate the tear samples contaminated with blood. Tear glucose monitoring might be a reliable and non-invasive substitute method for monitoring the blood glucose concentrations for diabetes patients, irrespective of glycated hemoglobin levels and timing of sample collection.


Subject(s)
Biomarkers/analysis , Blood Glucose/analysis , Diabetes Mellitus/diagnosis , Glycated Hemoglobin/analysis , Models, Statistical , Occult Blood , Tears/metabolism , Adult , Blood Glucose Self-Monitoring , Case-Control Studies , Diabetes Mellitus/epidemiology , Diabetes Mellitus/metabolism , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Tears/chemistry
6.
Nat Commun ; 9(1): 4863, 2018 11 19.
Article in English | MEDLINE | ID: mdl-30451856

ABSTRACT

M2a-subtype macrophage activation is known to be impaired in obesity, although the underlying mechanisms remain poorly understood. Herein, we demonstrate that, the IL-4/Irs2/Akt pathway is selectively impaired, along with decreased macrophage Irs2 expression, although IL-4/STAT6 pathway is maintained. Indeed, myeloid cell-specific Irs2-deficient mice show impairment of IL-4-induced M2a-subtype macrophage activation, as a result of stabilization of the FoxO1/HDAC3/NCoR1 corepressor complex, resulting in insulin resistance under the HF diet condition. Moreover, the reduction of macrophage Irs2 expression is mediated by hyperinsulinemia via the insulin receptor (IR). In myeloid cell-specific IR-deficient mice, the IL-4/Irs2 pathway is preserved in the macrophages, which results in a reduced degree of insulin resistance, because of the lack of IR-mediated downregulation of Irs2. We conclude that downregulation of Irs2 in macrophages caused by hyperinsulinemia is responsible for systemic insulin resistance via impairment of M2a-subtype macrophage activation in obesity.


Subject(s)
Hyperinsulinism/genetics , Insulin Receptor Substrate Proteins/genetics , Interleukin-4/genetics , Macrophages/metabolism , Obesity/genetics , 3T3-L1 Cells , Animals , Cell Movement , Cell Proliferation , Coculture Techniques , Diet, High-Fat/adverse effects , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Insulin Receptor Substrate Proteins/deficiency , Insulin Resistance , Interleukin-4/metabolism , Macrophage Activation , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Co-Repressor 1/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , STAT6 Transcription Factor/genetics , STAT6 Transcription Factor/metabolism , Signal Transduction
7.
Diabetes Technol Ther ; 19(11): 651-659, 2017 11.
Article in English | MEDLINE | ID: mdl-29024607

ABSTRACT

BACKGROUND: Performing self-monitoring of blood glucose (SMBG) is a clinical challenge for elderly people with low dexterity. An all-in-one-type SMBG device that can simply and easily puncture and monitor would be useful for them. We developed an automatic skin-puncturing and blood-sampling (APS) system for introducing of an all-in-one-type SMBG device. The aims of this study were to determine if the developed APS system, which has automatic puncturing, squeezing, and application functions, could provide sufficient blood sample volumes for SMBG and to determine the factors associated with failure in the use of the system by adult volunteers. METHODS: We investigated the success rate of obtaining a 0.8-µL sample volume using the APS system and determined the factors associated with failure in 140 adult volunteers. The participant characteristics, induration of puncturing sites, and states of finger grip conditions were evaluated as factors of a puncturing failure. The participant characteristics, skin hydration, states of finger grip, skin elasticity of the finger pad, and blood flow were evaluated as factors of a squeezing failure. RESULTS: The success rate was 61.9%. Puncturing failure was 21.6%, and squeezing failure was 16.5%. Automatic puncturing factors associated with failure were male sex, larger finger diameter, and thicker finger pad. The only squeezing failure factor was lower peripheral skin temperature. CONCLUSIONS: Improvement of the finger station groove shape to prevent ischemia and the squeezing angle would be useful developments of the all-in-one-type SMBG device for elderly people with decreased dexterity.


Subject(s)
Blood Glucose Self-Monitoring/instrumentation , Blood Glucose/analysis , Blood Specimen Collection/instrumentation , Adult , Female , Fingers , Humans , Male , Middle Aged , Skin
8.
Sci Rep ; 7(1): 5387, 2017 07 14.
Article in English | MEDLINE | ID: mdl-28710407

ABSTRACT

Several cellular signaling pathways, including insulin/IGF signaling, are known to be activated in hepatocellular carcinoma (HCC). Here, we investigated the roles of insulin receptor substrate (Irs) 1 and Irs2, both of which are the major molecules to be responsible for transducing insulin/IGF signaling in the liver, in the development of HCC by inducing chemical carcinogenesis using diethylnitrosamine (DEN) in mice. The Irs1 mRNA and protein expressions were upregulated in the tumors, along with enhanced insulin signaling. Liver-specific Irs1-knockout (LIrs1KO) mice exhibited suppression of DEN-induced HCC development, accompanied by reduced cancer cell proliferative activity and reduced activation of Akt. Gene expression analyses revealed that the tumors in the DEN-treated LIrs1KO mice showed modest metabolic alterations during hepatocarcinogenesis as well as decreased inflammation and invasion potentials. On the other hand, liver-specific Irs2-knockout (LIrs2KO) mice showed a similar pattern of HCC development to the DEN-treated control wild-type mice. Based on the knowledge that Wnt/ß-catenin signaling is activated in HCC, we focused on Wnt/ß-catenin signaling and demonstrated that Irs1 expression was induced by Wnt3a stimulation in the primary hepatocytes, associated with insulin-stimulated Akt activation. These data suggest that upregulated Irs1 by Wnt/ß-catenin signaling plays a crucial role in the progression of HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Insulin Receptor Substrate Proteins/genetics , Liver Neoplasms/genetics , Wnt3A Protein/genetics , beta Catenin/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Diethylnitrosamine , Disease Progression , Hepatocytes/metabolism , Hepatocytes/pathology , Insulin/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Liver/metabolism , Liver/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Wnt3A Protein/metabolism , beta Catenin/metabolism
9.
Diabetes ; 66(9): 2339-2350, 2017 09.
Article in English | MEDLINE | ID: mdl-28630133

ABSTRACT

Growing attention has been focused on the roles of the proximal tubules (PTs) of the kidney in glucose metabolism, including the mechanism of regulation of gluconeogenesis. In this study, we found that PT-specific insulin receptor substrate 1/2 double-knockout mice, established by using the newly generated sodium-glucose cotransporter 2 (SGLT2)-Cre transgenic mice, exhibited impaired insulin signaling and upregulated gluconeogenic gene expression and renal gluconeogenesis, resulting in systemic insulin resistance. In contrast, in streptozotocin-treated mice, although insulin action was impaired in the PTs, the gluconeogenic gene expression was unexpectedly downregulated in the renal cortex, which was restored by administration of an SGLT1/2 inhibitor. In the HK-2 cells, the gluconeogenic gene expression was suppressed by insulin, accompanied by phosphorylation and inactivation of forkhead box transcription factor 1 (FoxO1). In contrast, glucose deacetylated peroxisome proliferator-activated receptor γ coactivator 1-α (PGC1α), a coactivator of FoxO1, via sirtuin 1, suppressing the gluconeogenic gene expression, which was reversed by inhibition of glucose reabsorption. These data suggest that both insulin signaling and glucose reabsorption suppress the gluconeogenic gene expression by inactivation of FoxO1 and PGC1α, respectively, providing insight into novel mechanisms underlying the regulation of gluconeogenesis in the PTs.


Subject(s)
Gluconeogenesis/physiology , Glucose/metabolism , Insulin/metabolism , Kidney Tubules, Proximal/physiology , Animals , Cell Line , Diabetes Mellitus, Experimental/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/physiology , Humans , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Signal Transduction/physiology , Sirtuin 1/genetics , Sirtuin 1/metabolism , Sodium-Glucose Transporter 2/genetics , Sodium-Glucose Transporter 2/metabolism
10.
Stem Cells ; 35(7): 1835-1848, 2017 07.
Article in English | MEDLINE | ID: mdl-28480607

ABSTRACT

Myelotoxic injury, such as chemotherapeutic agents and ionizing radiation, unlocks the vigorous power of hematopoietic stem cells (HSCs) to replenish the hematopoietic system, making quiescent HSCs enter the cell cycle. Considering that both HSC-intrinsic and -extrinsic mechanisms enforce quiescence of HSCs, the drastic change in bone marrow (BM) environment after injury, represented by massive expansion of BM adipocytes, might trigger HSC activation. BM adipocytes, the major cellular component in the ablated marrow, however, reportedly suppress proliferation of hematopoietic cells, which may indicate the BM adipocytogenesis is an irrational response of injured organism. Given that adipose tissue is an endocrine organ with pleiotropic functions, we hypothesized that adipocyte-derived factors, especially adiponectin, an anti-inflammatory adipokine involved in regulation of granulopoiesis, are implicated in HSC activation. Myeloablative intervention increased BM adiponectin by multiple mechanisms, including adipocyte expansion and increased diffusion from the blood. Adiponectin-null (Adipoq -/- ) mice showed delayed hematopoietic recovery after BM injury, with Adipoq-/- HSCs more quiescent and defective in mammalian target of rapamycin complex 1 (mTORC1) activation. Recombinant adiponectin promoted not only HSC activation in vivo but cytokine-induced activation in vitro, and shortened the time for exit from quiescence in an mTORC1-dependent manner. These data illustrate a scarcely-reported example of a cell-extrinsic factor, adiponectin, enhancing quiescence exit of HSCs, and subsequent hematopoietic recovery. Our findings also highlight adipocytes as a source of adiponectin to ensure the proliferative burst of hematopoietic cells in ablated marrow. Stem Cells 2017;35:1835-1848.


Subject(s)
Adiponectin/genetics , Bone Marrow/metabolism , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Mechanistic Target of Rapamycin Complex 1/genetics , Adiponectin/deficiency , Animals , Benzhydryl Compounds/pharmacology , Bone Marrow/drug effects , Bone Marrow/pathology , Bone Marrow/radiation effects , Bone Marrow Transplantation , Cyclophosphamide/pharmacology , Cytarabine/pharmacology , Epoxy Compounds/pharmacology , Fluorouracil/pharmacology , Gene Expression Regulation , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/cytology , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Myeloablative Agonists/pharmacology , Poly I-C/pharmacology , Signal Transduction , Sirolimus/pharmacology , Whole-Body Irradiation
11.
Soc Sci Med ; 182: 45-51, 2017 06.
Article in English | MEDLINE | ID: mdl-28412640

ABSTRACT

BACKGROUND: Although living in a hilly environment may promote muscular activity in the daily lives of residents, and such activity may prevent diabetes mellitus, few studies have focused on the impact of living in a hilly environment on diabetes mellitus. The purpose of this study was to investigate the impact of a hilly neighborhood environment on DM in older people. METHODS: We used data from the Japan Gerontological Evaluation Study, a population-based, cross-sectional study of individuals aged 65 or older without long-term care needs in Japan, which was conducted in 2010. A total of 8904 participants in 46 neighborhoods had responded to the questionnaire and undergone a health check. Diabetes mellitus was diagnosed as HbA1c ≥ 6.5% and those undergoing treatment for diabetes mellitus. Poorly controlled diabetes mellitus was diagnosed in those without other chronic diseases who had an HbA1c > 7.5%, and in those with other chronic diseases if their HbA1c was >8.0%. Neighborhood environment was evaluated based on the percentage of positive responses in the questionnaire and geographical information system data. A multilevel analysis was performed, adjusted for individual-level risk factors. Furthermore, sensitivity analysis was conducted for those who were undergoing treatment for diabetes mellitus (n = 1007). RESULTS: After adjustment for other physical environmental and individual covariates, a 1 interquartile range increase (1.48°) in slope in the neighborhood decreased the risk of poorly controlled diabetes mellitus by 18% (odds ratio [OR]: 0.82, 95% confidence interval [CI]: 0.70-0.97). Sensitivity analysis confirmed that larger slopes in the neighborhood showed a significant protective effect against diabetes mellitus among those who were undergoing treatment for diabetes mellitus (OR: 0.73, 95% CI: 0.59-0.90). CONCLUSION: A hilly neighborhood environment was not associated with diabetes mellitus, but was protective against poorly controlled diabetes mellitus.


Subject(s)
Diabetes Mellitus/epidemiology , Environment , Geographic Mapping , Residence Characteristics/statistics & numerical data , Aged , Aged, 80 and over , Cross-Sectional Studies , Educational Status , Employment/methods , Employment/statistics & numerical data , Female , Glycated Hemoglobin/analysis , Humans , Income/statistics & numerical data , Japan/epidemiology , Male , Multilevel Analysis , Odds Ratio , Risk Factors
12.
Nat Commun ; 7: 12977, 2016 10 06.
Article in English | MEDLINE | ID: mdl-27708333

ABSTRACT

Hepatic insulin signalling involves insulin receptor substrates (Irs) 1/2, and is normally associated with the inhibition of gluconeogenesis and activation of lipogenesis. In diabetes and obesity, insulin no longer suppresses hepatic gluconeogenesis, while continuing to activate lipogenesis, a state referred to as 'selective insulin resistance'. Here, we show that 'selective insulin resistance' is caused by the differential expression of Irs1 and Irs2 in different zones of the liver. We demonstrate that hepatic Irs2-knockout mice develop 'selective insulin resistance', whereas mice lacking in Irs1, or both Irs1 and Irs2, develop 'total insulin resistance'. In obese diabetic mice, Irs1/2-mediated insulin signalling is impaired in the periportal zone, which is the primary site of gluconeogenesis, but enhanced in the perivenous zone, which is the primary site of lipogenesis. While hyperinsulinaemia reduces Irs2 expression in both the periportal and perivenous zones, Irs1 expression, which is predominantly in the perivenous zone, remains mostly unaffected. These data suggest that 'selective insulin resistance' is induced by the differential distribution, and alterations of hepatic Irs1 and Irs2 expression.


Subject(s)
Antigens, CD/metabolism , Diabetes Mellitus/metabolism , Insulin Resistance , Liver/metabolism , Obesity/metabolism , Receptor, Insulin/metabolism , Animals , Cell Nucleus/metabolism , Diabetes Mellitus, Experimental/metabolism , Gluconeogenesis , Homeostasis , Humans , Hyperinsulinism/metabolism , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/metabolism , Signal Transduction
13.
Sci Rep ; 6: 34707, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703271

ABSTRACT

The aim of this study is to elucidate to what degree adiponectin is involved in TZD-mediated amelioration of neointimal formation. We investigated the effect of 3- or 8-weeks' pioglitazone on cuff-induced neointimal formation in adiponectin-deficient (APN-KO) and wild-type (WT) mice. Pioglitazone for 3 weeks reduced neointimal formation in the WT mice with upregulation of the plasma adiponectin levels, but failed to reduce neointimal formation in the APN-KO mice, suggesting that pioglitazone suppressed neointimal formation by adiponectin-dependent mechanisms. Pioglitazone for 3 weeks suppressed vascular smooth muscle cell (VSMC) proliferation and increased AdipoR2 expression in the WT mice. In vitro, globular adiponectin activated AMPK through both AdipoR1 and AdipoR2, resulting in the inhibition of VSMC proliferation. Interestingly, 8-weeks' pioglitazone was reduced neointimal formation in APN-KO mice to degree similar to that seen in the WT mice, suggesting that pioglitazone can also suppress neointimal formation via a mechanism independent of adiponectin. Pioglitazone for 8 weeks completely abrogated the increased VSMC proliferation, along with a reduction of cyclin B1 and cyclin D1 expressions and cardiovascular risk profile in the APN-KO mice. In vitro, pioglitazone suppressed these expressions, leading to inhibition of VSMC proliferation. Pioglitazone suppresses neointimal formation via both adiponectin-dependent and adiponectin-independent mechanisms.


Subject(s)
Adiponectin/blood , Adiponectin/genetics , Muscle, Smooth, Vascular/cytology , Neointima/drug therapy , Thiazolidinediones/administration & dosage , Animals , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Humans , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Neointima/etiology , Neointima/genetics , Neointima/metabolism , Pioglitazone , Receptors, Adiponectin/metabolism , Thiazolidinediones/pharmacology
14.
Diabetologia ; 59(11): 2426-2434, 2016 11.
Article in English | MEDLINE | ID: mdl-27525648

ABSTRACT

AIMS/HYPOTHESIS: Recently, incretin-related agents have been reported to attenuate insulin resistance in animal models, although the underlying mechanisms remain unclear. In this study, we investigated whether anagliptin, the dipeptidyl peptidase 4 (DPP-4) inhibitor, attenuates skeletal muscle insulin resistance through endothelial nitric oxide synthase (eNOS) activation in the endothelial cells. We used endothelium-specific Irs2-knockout (ETIrs2KO) mice, which show skeletal muscle insulin resistance resulting from a reduction of insulin-induced skeletal muscle capillary recruitment as a consequence of impaired eNOS activation. METHODS: In vivo, 8-week-old male ETIrs2KO mice were fed regular chow with or without 0.3% (wt/wt) DPP-4 inhibitor for 8 weeks to assess capillary recruitment and glucose uptake by the skeletal muscle. In vitro, human coronary arterial endothelial cells (HCAECs) were used to explore the effect of glucagon-like peptide 1 (GLP-1) on eNOS activity. RESULTS: Treatment with anagliptin ameliorated the impaired insulin-induced increase in capillary blood volume, interstitial insulin concentration and skeletal muscle glucose uptake in ETIrs2KO mice. This improvement in insulin-induced glucose uptake was almost completely abrogated by the GLP-1 receptor (GLP-1R) antagonist exendin-(9-39). Moreover, the increase in capillary blood volume with anagliptin treatment was also completely inhibited by the NOS inhibitor. GLP-1 augmented eNOS phosphorylation in HCAECs, with the effect completely disappearing after exposure to the protein kinase A (PKA) inhibitor H89. These data suggest that anagliptin treatment enhances insulin-induced capillary recruitment and interstitial insulin concentrations, resulting in improved skeletal muscle glucose uptake by directly acting on the endothelial cells via NO- and GLP-1-dependent mechanisms in vivo. CONCLUSIONS/INTERPRETATION: Anagliptin may be a promising agent to ameliorate skeletal muscle insulin resistance in obese patients with type 2 diabetes.


Subject(s)
Insulin/pharmacology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitrogen Oxides/metabolism , Pyrimidines/pharmacology , Animals , Dipeptidyl Peptidase 4/blood , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Insulin Receptor Substrate Proteins/deficiency , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , Male , Mice , Nitric Oxide Synthase Type III/metabolism , Pyrimidines/blood , Tandem Mass Spectrometry
15.
Immunity ; 44(6): 1422-33, 2016 06 21.
Article in English | MEDLINE | ID: mdl-27317261

ABSTRACT

Obesity has been shown to increase the morbidity of infections, however, the underlying mechanisms remain largely unknown. Here we demonstrate that obesity caused adiponectin deficiency in the bone marrow (BM), which led to an inflamed BM characterized by increased tumor necrosis factor (TNF) production from bone marrow macrophages. Hematopoietic stem and progenitor cells (HSPCs) chronically exposed to excessive TNF in obese marrow aberrantly expressed cytokine signaling suppressor SOCS3, impairing JAK-STAT mediated signal transduction and cytokine-driven cell proliferation. Accordingly, both obese and adiponectin-deficient mice showed attenuated clearance of infected Listeria monocytogenes, indicating that obesity or loss of adiponectin is critical for exacerbation of infection. Adiponectin treatment restored the defective HSPC proliferation and bacterial clearance of obese and adiponectin-deficient mice, affirming the importance of adiponectin against infection. Taken together, our findings demonstrate that obesity impairs hematopoietic response against infections through a TNF-SOCS3-STAT3 axis, highlighting adiponectin as a legitimate target against obesity-related infections.


Subject(s)
Adiponectin/metabolism , Hematopoietic Stem Cells/physiology , Inflammation/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Obesity/immunology , Adiponectin/genetics , Animals , Bacteriolysis , Bone Marrow/immunology , Cells, Cultured , Diet , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cell Mobilization , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Tumor Necrosis Factor-alpha/metabolism
17.
Endocrinology ; 157(3): 1029-42, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26713783

ABSTRACT

Sodium glucose cotransporter 2 inhibitors have attracted attention as they exert antidiabetic and antiobesity effects. In this study, we investigated the effects of tofogliflozin on glucose homeostasis and its metabolic consequences and clarified the underlying molecular mechanisms. C57BL/6 mice were fed normal chow containing tofogliflozin (0.005%) for 20 weeks or a high-fat diet containing tofogliflozin (0.005%) for 8 weeks ad libitum. In addition, the animals were pair-fed in relation to controls to exclude the influence of increased food intake. Tofogliflozin reduced the body weight gain, mainly because of fat mass reduction associated with a diminished adipocyte size. Glucose tolerance and insulin sensitivity were ameliorated. The serum levels of nonesterified fatty acid and ketone bodies were increased and the respiratory quotient was decreased in the tofogliflozin-treated mice, suggesting the acceleration of lipolysis in the white adipose tissue and hepatic ß-oxidation. In fact, the phosphorylation of hormone-sensitive lipase and the adipose triglyceride lipase protein levels in the white adipose tissue as well as the gene expressions related to ß-oxidation, such as Cpt1α in the liver, were significantly increased. The hepatic triglyceride contents and the expression levels of lipogenic genes were decreased. Pair-fed mice exhibited almost the same results as mice fed an high-fat diet ad libitum. Moreover, a hyperinsulinemic-euglycemic clamp revealed that tofogliflozin improved insulin resistance by increasing glucose uptake, especially in the skeletal muscle, in pair-fed mice. Taken together, these results suggest tofogliflozin ameliorates insulin resistance and obesity by increasing glucose uptake in skeletal muscle and lipolysis in adipose tissue.


Subject(s)
Adipose Tissue, White/drug effects , Benzhydryl Compounds/pharmacology , Glucosides/pharmacology , Hypoglycemic Agents/pharmacology , Insulin Resistance , Lipolysis/drug effects , Muscle, Skeletal/drug effects , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Adipose Tissue, White/metabolism , Animals , Diet, High-Fat , Fatty Acids, Nonesterified/blood , Gene Expression/drug effects , Glucose/metabolism , Glucose Clamp Technique , Ketone Bodies/blood , Lipase/drug effects , Lipase/metabolism , Lipid Metabolism/drug effects , Lipogenesis/drug effects , Lipogenesis/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Sodium-Glucose Transporter 2 Inhibitors , Sterol Esterase/drug effects , Sterol Esterase/metabolism , Weight Gain/drug effects
18.
Proc Natl Acad Sci U S A ; 112(10): E1067-76, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25713368

ABSTRACT

Increase in the concentration of plasma L-cysteine is closely associated with defective insulin secretion from pancreatic ß-cells, which results in type 2 diabetes (T2D). In this study, we investigated the effects of prolonged L-cysteine treatment on glucose-stimulated insulin secretion (GSIS) from mouse insulinoma 6 (MIN6) cells and from mouse pancreatic islets, and found that the treatment reversibly inhibited glucose-induced ATP production and resulting GSIS without affecting proinsulin and insulin synthesis. Comprehensive metabolic analyses using capillary electrophoresis time-of-flight mass spectrometry showed that prolonged L-cysteine treatment decreased the levels of pyruvate and its downstream metabolites. In addition, methyl pyruvate, a membrane-permeable form of pyruvate, rescued L-cysteine-induced inhibition of GSIS. Based on these results, we found that both in vitro and in MIN6 cells, L-cysteine specifically inhibited the activity of pyruvate kinase muscle isoform 2 (PKM2), an isoform of pyruvate kinases that catalyze the conversion of phosphoenolpyruvate to pyruvate. L-cysteine also induced PKM2 subunit dissociation (tetramers to dimers/monomers) in cells, which resulted in impaired glucose-induced ATP production for GSIS. DASA-10 (NCGC00181061, a substituted N,N'-diarylsulfonamide), a specific activator for PKM2, restored the tetramer formation and the activity of PKM2, glucose-induced ATP production, and biphasic insulin secretion in L-cysteine-treated cells. Collectively, our results demonstrate that impaired insulin secretion due to exposure to L-cysteine resulted from its direct binding and inactivation of PKM2 and suggest that PKM2 is a potential therapeutic target for T2D.


Subject(s)
Adenosine Triphosphate/biosynthesis , Carrier Proteins/antagonists & inhibitors , Cysteine/pharmacology , Glucose/pharmacology , Insulin/metabolism , Membrane Proteins/antagonists & inhibitors , Animals , Cell Line , Insulin Secretion , Mice , Thyroid Hormones , Thyroid Hormone-Binding Proteins
19.
Diabetes ; 64(3): 876-86, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25277391

ABSTRACT

Endothelial cells are considered to be essential for normal pancreatic ß-cell function. The current study attempted to demonstrate the role of insulin receptor substrate-2 (Irs2) in endothelial cells with regard to insulin secretion. Endothelial cell-specific Irs2 knockout (ETIrs2KO) mice exhibited impaired glucose-induced, arginine-induced, and glucagon-induced insulin secretion and showed glucose intolerance. In batch incubation and perifusion experiments using isolated islets, glucose-induced insulin secretion was not significantly different between the control and the ETIrs2KO mice. In contrast, in perfusion experiments, glucose-induced insulin secretion was significantly impaired in the ETIrs2KO mice. The islet blood flow was significantly impaired in the ETIrs2KO mice. After the treatment of these knockout mice with enalapril maleate, which improved the islet blood flow, glucose-stimulated insulin secretion was almost completely restored to levels equal to those in the control mice. These data suggest that Irs2 deletion in endothelial cells leads to a decreased islet blood flow, which may cause impaired glucose-induced insulin secretion. Thus, Irs2 in endothelial cells may serve as a novel therapeutic target for preventing and ameliorating type 2 diabetes and metabolic syndrome.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Insulin Receptor Substrate Proteins/metabolism , Insulin/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Enalapril/pharmacology , Insulin Receptor Substrate Proteins/genetics , Insulin Secretion , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Knockout
20.
Diabetologia ; 57(3): 542-53, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24317852

ABSTRACT

AIMS/HYPOTHESIS: Common genetic variations of the transcription factor 7-like 2 gene (encoded by TCF7L2), one of the T cell factor/lymphoid enhancer-binding factor transcription factors for the converging wingless-type MMTV integration site family (Wnt)/ß-catenin signalling pathway, are known to be associated with type 2 diabetes. Individuals with at-risk alleles of TCF7L2 exhibit impaired insulin secretion. Although previous studies using animal models have revealed the existence of a relationship between the Wnt/ß-catenin signalling pathway and glucose homeostasis, it remains unclear whether TCF7L2 in the pancreatic beta cells might be causally involved in insulin secretion in vivo. In this study, we investigated the role of TCF7L2 expressed in the pancreatic beta cells in glucose homeostasis. METHODS: Three independent groups of genetically engineered mice (DN mice) were generated, in which expression of the dominant-negative form of Tcf7l2 was driven under a rat insulin promoter. Phenotypes of both adult and newborn mice were evaluated. The levels of genes and proteins expressed in isolated islets were determined by reverse transcription-quantitative PCR and western blot analysis, respectively. RESULTS: Adult DN mice showed impaired glucose tolerance and decreased insulin secretion in both oral and intraperitoneal glucose tolerance tests. Marked reduction of the beta cell area and whole-pancreas insulin content was observed in both the adult and newborn DN mice. Islets from the DN mice showed decreased gene expressions of Ccnd1, Ccnd2, Irs1, Irs2, Ins1, Ins2 and Mafa, consistent with the deleterious effects of the dominant-negative form of Tcf7l2 on beta cell proliferation and insulin production. CONCLUSIONS/INTERPRETATION: TCF7L2 expressed in the pancreatic beta cells plays a crucial role in glucose metabolism through regulation of the beta cell mass.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , Pancreas/metabolism , Transcription Factor 7-Like 2 Protein/metabolism , Animals , Blotting, Western , Cells, Cultured , Gene Expression Regulation , Homeostasis , Insulin Secretion , Insulin-Secreting Cells/metabolism , Islets of Langerhans/cytology , Mice , Pancreas/cytology , Reverse Transcriptase Polymerase Chain Reaction , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein/genetics , Wnt Signaling Pathway
SELECTION OF CITATIONS
SEARCH DETAIL
...