Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Diabetes Investig ; 15(4): 437-448, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38151917

ABSTRACT

AIMS/INTRODUCTION: Endogenous insulin secretion could be recovered by improving hyperglycemia in patients with type 2 diabetes. This study aimed to investigate the association between short-term recovery of insulin secretion during hospitalization and clinical background or future glycemic control in patients with type 2 diabetes. MATERIALS AND METHODS: A total of 127 patients with type 2 diabetes were included. The recovery of endogenous insulin secretion was determined using the following indices: index A: fasting C-peptide index (CPI) at discharge - fasting CPI on admission; index B: postprandial CPI at discharge - postprandial CPI on admission; and index C: Δ C-peptide immunoreactivity (CPR) (postprandial CPR - fasting CPR) at discharge - ΔCPR on admission. We examined the associations of each index with clinical background and future glycemic control measured by glycosylated hemoglobin and continuous glucose monitoring. RESULTS: Using index A and B, the age was significantly younger, whereas BMI and visceral fat area were significantly higher in the high-recovery group than in the low-recovery group. Changes in glycosylated hemoglobin levels were significantly greater at 6 and 12 months in the high-recovery group in the analysis of index C. The receiver operating characteristic curve analysis identified the index B and index C as indicators to predict glycosylated hemoglobin <7.0% at 6 months after discharge. Furthermore, index C was positively correlated with the time in the target glucose range, and inversely correlated with the standard deviation of glucose at 3 and 12 months after discharge. CONCLUSIONS: Short-term recovery of insulin secretion in response to a meal during hospitalization, evaluated with the index-C, might predict future glycemic control.


Subject(s)
Diabetes Mellitus, Type 2 , Humans , Insulin Secretion , Insulin/metabolism , Glycated Hemoglobin , C-Peptide/metabolism , Blood Glucose/analysis , Blood Glucose Self-Monitoring , Glycemic Control , Glucose , Postprandial Period/physiology
2.
Mar Drugs ; 21(10)2023 Sep 28.
Article in English | MEDLINE | ID: mdl-37888449

ABSTRACT

Astaxanthin (AX), a lipid-soluble pigment belonging to the xanthophyll carotenoids family, has recently garnered significant attention due to its unique physical properties, biochemical attributes, and physiological effects. Originally recognized primarily for its role in imparting the characteristic red-pink color to various organisms, AX is currently experiencing a surge in interest and research. The growing body of literature in this field predominantly focuses on AXs distinctive bioactivities and properties. However, the potential of algae-derived AX as a solution to various global environmental and societal challenges that threaten life on our planet has not received extensive attention. Furthermore, the historical context and the role of AX in nature, as well as its significance in diverse cultures and traditional health practices, have not been comprehensively explored in previous works. This review article embarks on a comprehensive journey through the history leading up to the present, offering insights into the discovery of AX, its chemical and physical attributes, distribution in organisms, and biosynthesis. Additionally, it delves into the intricate realm of health benefits, biofunctional characteristics, and the current market status of AX. By encompassing these multifaceted aspects, this review aims to provide readers with a more profound understanding and a robust foundation for future scientific endeavors directed at addressing societal needs for sustainable nutritional and medicinal solutions. An updated summary of AXs health benefits, its present market status, and potential future applications are also included for a well-rounded perspective.


Subject(s)
Antioxidants , Xanthophylls , Xanthophylls/chemistry
3.
J Diabetes Investig ; 13(6): 1052-1061, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35092353

ABSTRACT

AIMS/INTRODUCTION: Diastolic cardiac dysfunction in type 2 diabetes (DD2D) is a critical risk of heart failure with preserved ejection fraction. However, there is no established biomarker to detect DD2D. We aimed to investigate the predictive impact of fragmented QRS (fQRS) on electrocardiography on the existence of DD2D. MATERIALS AND METHODS: We included in-hospital patients with type 2 diabetes without heart failure symptoms who were admitted to our institution for glycemic management between November 2017 and April 2021. An fQRS was defined as an additional R' wave or notching/splitting of the S wave in two contiguous electrocardiography leads. DD2D was diagnosed according to the latest guidelines of the American Society of Echocardiography. RESULTS: Of 320 participants, 122 patients (38.1%) had fQRS. DD2D was diagnosed in 82 (25.6%). An fQRS was significantly associated with the existence of DD2D (odds ratio 4.37, 95% confidence interval 2.33-8.20; p < 0.0001) adjusted for seven potential confounders. The correlation between DD2D and diabetic microvascular disease was significant only among those with fQRS. Classification and regression tree analysis showed that fQRS was the most relevant optimum split for DD2D. CONCLUSIONS: An fQRS might be a simple and promising predictor of the existence of DD2D. The findings should be validated in a larger-scale cohort.


Subject(s)
Diabetes Mellitus, Type 2 , Heart Diseases , Heart Failure , Diabetes Mellitus, Type 2/complications , Electrocardiography , Heart , Humans
4.
Nutrients ; 13(12)2021 Dec 06.
Article in English | MEDLINE | ID: mdl-34959926

ABSTRACT

Recently, obesity-induced insulin resistance, type 2 diabetes, and cardiovascular disease have become major social problems. We have previously shown that Astaxanthin (AX), which is a natural antioxidant, significantly ameliorates obesity-induced glucose intolerance and insulin resistance. It is well known that AX is a strong lipophilic antioxidant and has been shown to be beneficial for acute inflammation. However, the actual effects of AX on chronic inflammation in adipose tissue (AT) remain unclear. To observe the effects of AX on AT functions in obese mice, we fed six-week-old male C57BL/6J on high-fat-diet (HFD) supplemented with or without 0.02% of AX for 24 weeks. We determined the effect of AX at 10 and 24 weeks of HFD with or without AX on various parameters including insulin sensitivity, glucose tolerance, inflammation, and mitochondrial function in AT. We found that AX significantly reduced oxidative stress and macrophage infiltration into AT, as well as maintaining healthy AT function. Furthermore, AX prevented pathological AT remodeling probably caused by hypoxia in AT. Collectively, AX treatment exerted anti-inflammatory effects via its antioxidant activity in AT, maintained the vascular structure of AT and preserved the stem cells and progenitor's niche, and enhanced anti-inflammatory hypoxia induction factor-2α-dominant hypoxic response. Through these mechanisms of action, it prevented the pathological remodeling of AT and maintained its integrity.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/physiology , Anti-Inflammatory Agents , Antioxidants , Dietary Supplements , Adipose Tissue/pathology , Animals , Cytokines/metabolism , Glucose/metabolism , Inflammation , Inflammation Mediators/metabolism , Insulin Resistance , Macrophages/pathology , Male , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/physiology , Oxidative Stress/drug effects , Xanthophylls/administration & dosage , Xanthophylls/pharmacology
5.
J Diabetes Res ; 2021: 8838026, 2021.
Article in English | MEDLINE | ID: mdl-33855087

ABSTRACT

OBJECTIVES: A single-arm prospective study was conducted among Japanese patients with type 2 diabetes having preserved ejection fraction. The aim was to investigate (1) whether liraglutide therapy could improve B-type natriuretic peptide (BNP) levels and diastolic cardiac function assessed by the E-wave to E' ratio (E/E') using transthoracic echocardiography (TTE), and (2) whether E/E' contributed to BNP improvement independent of bodyweight reduction (UMIN000005565). METHODS: Patients with type 2 diabetes and left ventricular ejection fraction (LVEF) ≥ 40% without heart failure symptoms were enrolled, and daily injection with liraglutide (0.9 mg) was introduced. Cardiac functions were assessed by TTE before and after 26 weeks of liraglutide treatment. Diastolic cardiac function was defined as septal E/E' ≥ 13.0. RESULTS: Thirty-one patients were analyzed. BNP and E/E' improved, with BNP levels declining from 36.8 ± 30.5 pg/mL to 26.3 ± 25.9 pg/mL (p = 0.0014) and E/E' dropping from 12.7 ± 4.7 to 11.0 ± 3.3 (p = 0.0376). The LVEF showed no significant changes. E/E' improved only in patients with E/E' ≥ 13.0. Favorable changes in E/E' were canceled when adjusted for body mass index (BMI). Multivariate linear regression analysis revealed that the left ventricular diastolic diameter and ∆E/E'/∆BMI contributed to ∆BNP/baseline BNP (p = 0.0075, R 2 = 0.49264). CONCLUSIONS: Liraglutide had favorable effects on BNP and E/E' but not on LVEF. E/E' improvement was only seen in patients with diastolic cardiac function. Body weight reduction affected the change of E/E'. The BMI-adjusted E/E' significantly contributed to the relative change of BNP. GLP-1 analog treatment could be considered a therapeutic option against diabetic diastolic cardiac dysfunction regardless of body weight. This trial is registered with the University Hospital Medical Information Network in Japan, with clinical trial registration number: UMIN000005565.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Diastole/drug effects , Liraglutide/therapeutic use , Natriuretic Peptide, Brain/blood , Stroke Volume/physiology , Weight Loss/drug effects , Aged , Diabetes Mellitus, Type 2/physiopathology , Diastole/physiology , Female , Humans , Liraglutide/pharmacology , Male , Middle Aged
6.
J Cachexia Sarcopenia Muscle ; 11(1): 241-258, 2020 02.
Article in English | MEDLINE | ID: mdl-32003547

ABSTRACT

BACKGROUND: Skeletal muscle is mainly responsible for insulin-stimulated glucose disposal. Dysfunction in skeletal muscle metabolism especially during obesity contributes to the insulin resistance. Astaxanthin (AX), a natural antioxidant, has been shown to ameliorate hepatic insulin resistance in obese mice. However, its effects in skeletal muscle are poorly understood. The current study aimed to investigate the molecular target of AX in ameliorating skeletal muscle insulin resistance. METHODS: We fed 6-week-old male C57BL/6J mice with normal chow (NC) or NC supplemented with AX (NC+AX) and high-fat-diet (HFD) or HFD supplemented with AX for 24 weeks. We determined the effect of AX on various parameters including insulin sensitivity, glucose uptake, inflammation, kinase signaling, gene expression, and mitochondrial function in muscle. We also determined energy metabolism in intact C2C12 cells treated with AX using the Seahorse XFe96 Extracellular Flux Analyzer and assessed the effect of AX on mitochondrial oxidative phosphorylation and mitochondrial biogenesis. RESULTS: AX-treated HFD mice showed improved metabolic status with significant reduction in blood glucose, serum total triglycerides, and cholesterol (p< 0.05). AX-treated HFD mice also showed improved glucose metabolism by enhancing glucose incorporation into peripheral target tissues, such as the skeletal muscle, rather than by suppressing gluconeogenesis in the liver as shown by hyperinsulinemic-euglycemic clamp study. AX activated AMPK in the skeletal muscle of the HFD mice and upregulated the expressions of transcriptional factors and coactivator, thereby inducing mitochondrial remodeling, including increased mitochondrial oxidative phosphorylation component and free fatty acid metabolism. We also assessed the effects of AX on mitochondrial biogenesis in the siRNA-mediated AMPK-depleted C2C12 cells and showed that the effect of AX was lost in the genetically AMPK-depleted C2C12 cells. Collectively, AX treatment (i) significantly ameliorated insulin resistance and glucose intolerance through regulation of AMPK activation in the muscle, (ii) stimulated mitochondrial biogenesis in the muscle, (iii) enhanced exercise tolerance and exercise-induced fatty acid metabolism, and (iv) exerted antiinflammatory effects via its antioxidant activity in adipose tissue. CONCLUSIONS: We concluded that AX treatment stimulated mitochondrial biogenesis and significantly ameliorated insulin resistance through activation of AMPK pathway in the skeletal muscle.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fibrinolytic Agents/therapeutic use , Insulin Resistance/physiology , Mitochondria, Muscle/metabolism , Animals , Fibrinolytic Agents/pharmacology , Humans , Male , Mice , Organelle Biogenesis , Xanthophylls/pharmacology , Xanthophylls/therapeutic use
7.
Sci Rep ; 9(1): 12284, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31439906

ABSTRACT

Obesity increases the risk of cancer. Increased levels of hormones (such as oestrogen, insulin, insulin-like growth factor, and leptin), free fatty acid-induced production of reactive oxygen species, an altered intestinal microbiome and chronic inflammation are known to be associated with an increased cancer risk in obese subjects. However, the mechanism underlying the connection between obesity and cancer development remains elusive. Here, we show that a high-fat diet (HFD) promotes tumour initiation/progression and induces a phenotypic switch from PD-1- CD8+ non-exhausted T cells to PD-1+ CD8+ exhausted T cells in a murine breast cancer model. While PD-1- CD8+ non-exhausted T cells predominated in the mammary glands of normal diet (ND)-fed mice, PD-1+ CD8+ exhausted T cells accumulated in the developing tumours of HFD-fed mice. Gene expression profiles indicated that PD-1+ CD8+ T cells expressed higher levels of the tumour-trophic gene Opn and lower levels of the cytotoxic genes Ifng and Gzmb than did PD-1- CD8+ T cells. Our study provides a possible mechanistic linkage between obesity and cancer.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Carcinogenesis , Dietary Fats/adverse effects , Mammary Neoplasms, Experimental , Obesity , Animals , CD8-Positive T-Lymphocytes/pathology , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/immunology , Carcinogenesis/pathology , Dietary Fats/pharmacology , Female , Mammary Neoplasms, Experimental/chemically induced , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/pathology , Mice , Obesity/chemically induced , Obesity/genetics , Obesity/immunology , Obesity/pathology , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology
8.
Endocr J ; 66(12): 1113-1116, 2019 Dec 25.
Article in English | MEDLINE | ID: mdl-31391355

ABSTRACT

We report a sporadic case of maturity-onset diabetes of the young type 5 (MODY5) with a whole-gene deletion of the hepatocyte nuclear factor-1beta (HNF1B) gene. A 44-year-old Japanese man who had been diagnosed with early-onset non-autoimmune diabetes mellitus at the age of 23 was examined. He showed multi-systemic symptoms, including a solitary congenital kidney, pancreatic hypoplasia, pancreatic exocrine dysfunction, elevation of the serum levels of liver enzymes, hypomagnesemia, and hyperuricemia. These clinical characteristics, in spite of the absence of a family history of diabetes, prompted us to make the diagnosis of maturity-onset diabetes of the young 5 (MODY 5). One allele deletion of the entire HNF1B gene revealed by multiplex ligation-dependent probe amplification (MLPA) led us to the diagnoses of 17q12 microdeletion syndrome even though there were negative chromosomal analyses with array comparative genomic hybridization (CGH). 17q12 microdeletion syndrome, which is not rare especially in sporadic cases since 17q12 is a typical hot spot for chromosomal deletion, could have complicated the clinical heterogeneity of MODY5.


Subject(s)
Chromosomes, Human, Pair 17/genetics , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/genetics , Gene Deletion , Hepatocyte Nuclear Factor 1-beta/genetics , Adult , Calcium/urine , Diabetes Mellitus, Type 2/physiopathology , Humans , Japan , Liver/enzymology , Magnesium/blood , Male , Pancreas/physiopathology , Syndrome , Tomography, X-Ray Computed
9.
J Diabetes Investig ; 10(6): 1411-1418, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30897274

ABSTRACT

AIMS/INTRODUCTION: Chronic inflammation of the liver is often observed with obesity or type 2 diabetes. In these pathological conditions, the immunological cells, such as macrophages, play important roles in the development or growth of liver cancer. Recently, it was reported that hypoxia-inducible factor-1α (HIF-1α) is a key molecule for the acquisition of inflammatory M1 polarity of macrophages. In the present study, we examined the effects of altered macrophage polarity on obesity- and diabetes-associated liver cancer using macrophage-specific HIF-1α knockout (KO) mice. MATERIALS AND METHODS: To induce liver cancer in the mice, diethylnitrosamine, a chemical carcinogen, was used. Both KO mice and wild-type littermates were fed either a high-fat diet (HFD) or normal chow. They were mainly analyzed 6 months after HFD feeding. RESULTS: Development of liver cancer after HFD feeding was 45% less in KO mice than in wild-type littermates mice. Phosphorylation of extracellular signal-regulated kinase 2 was also lower in the liver of KO mice. Those effects of HIF-1α deletion in macrophages were not observed in normal chow-fed mice. Furthermore, the size of liver tumors did not differ between KO and wild-type littermates mice, even those on a HFD. These results suggest that the activation of macrophage HIF-1α by HFD is involved not in the growth, but in the development of liver cancer with the enhanced oncogenic extracellular signal-regulated kinase 2 signaling in hepatocytes. CONCLUSIONS: The activation of macrophage HIF-1α might play important roles in the development of liver cancer associated with diet-induced obesity and diabetes.


Subject(s)
Diabetes Mellitus, Experimental/complications , Diet, High-Fat , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Inflammation/prevention & control , Liver Neoplasms/prevention & control , Macrophages/metabolism , Obesity/complications , Animals , Diabetes Mellitus, Experimental/physiopathology , Inflammation/etiology , Inflammation/pathology , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/physiopathology
10.
Sci Rep ; 8(1): 14597, 2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30254249

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

11.
Sci Rep ; 8(1): 11370, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054532

ABSTRACT

Sirt1 plays an important role in regulating glucose and lipid metabolism in obese animal models. Impaired adipose tissue angiogenesis in the obese state decreases adipogenesis and thereby contributes to glucose intolerance and lipid metabolism. However, the mechanism by which Sirt1 activation affects obesity-associated impairments in angiogenesis in the adipose tissue is not fully understood. Here, we show that SRT1720 treatment induces angiogenic genes in cultured 3T3-L1 preadipocytes and ex vivo preadipocytes. siRNA-mediated knockdown of Sirt1 in 3T3-L1 preadipocytes downregulated angiogenic genes in the preadipocytes. SRT1720 treatment upregulated metabolically favorable genes and reduced inflammatory gene expressions in the adipose tissue of diet-induced obese (DIO) mice. Collectively, these findings suggest a novel role of SRT1720-induced Sirt1 activation in the induction of angiogenic genes in preadipocytes, thereby reducing inflammation and fibrosis in white adipose tissue (WAT) and promoting insulin sensitivity.


Subject(s)
Adipocytes/metabolism , Diet, High-Fat , Gene Expression Regulation , Insulin Resistance/genetics , Neovascularization, Physiologic/genetics , Sirtuin 1/metabolism , 3T3-L1 Cells , Adipocytes/drug effects , Adipose Tissue, White/metabolism , Animals , Fibrosis , Gene Expression Regulation/drug effects , Glucose/metabolism , Heterocyclic Compounds, 4 or More Rings/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Neovascularization, Physiologic/drug effects
12.
Nat Commun ; 8(1): 286, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28819169

ABSTRACT

Adipose tissue resident macrophages have important roles in the maintenance of tissue homeostasis and regulate insulin sensitivity for example by secreting pro-inflammatory or anti-inflammatory cytokines. Here, we show that M2-like macrophages in adipose tissue regulate systemic glucose homeostasis by inhibiting adipocyte progenitor proliferation via the CD206/TGFß signaling pathway. We show that adipose tissue CD206+ cells are primarily M2-like macrophages, and ablation of CD206+ M2-like macrophages improves systemic insulin sensitivity, which was associated with an increased number of smaller adipocytes. Mice genetically engineered to have reduced numbers of CD206+ M2-like macrophages show a down-regulation of TGFß signaling in adipose tissue, together with up-regulated proliferation and differentiation of adipocyte progenitors. Our findings indicate that CD206+ M2-like macrophages in adipose tissues create a microenvironment that inhibits growth and differentiation of adipocyte progenitors and, thereby, control adiposity and systemic insulin sensitivity.Adipose tissue contains macrophages that can influence both local and systemic metabolism via the secretion of cytokines. Here, Nawaz et al. report that M2-like macrophages, present in adipose tissue, create a microenvironment that inhibits proliferation of adipocyte progenitors due to the secretion of TGF-ß1.


Subject(s)
Adipocytes/cytology , Glucose/metabolism , Lectins, C-Type/metabolism , Macrophages/metabolism , Mannose-Binding Lectins/metabolism , Obesity/metabolism , Receptors, Cell Surface/metabolism , Adipocytes/metabolism , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adipose Tissue, White/cytology , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Cell Proliferation , Diet, High-Fat/adverse effects , Insulin Resistance , Lectins, C-Type/genetics , Mannose Receptor , Mannose-Binding Lectins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Cell Surface/genetics , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism
13.
Diabetes ; 65(12): 3649-3659, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27625023

ABSTRACT

Adipose tissue hypoxia is an important feature of pathological adipose tissue expansion. Hypoxia-inducible factor-1α (HIF-1α) in adipocytes reportedly induces oxidative stress and fibrosis, rather than neoangiogenesis via vascular endothelial growth factor (VEGF)-A. We previously reported that macrophages in crown-like structures (CLSs) are both hypoxic and inflammatory. In the current study, we examined how macrophage HIF-1α is involved in high-fat diet (HFD)-induced inflammation, neovascularization, hypoxia, and insulin resistance using mice with myeloid cell-specific HIF-1α deletion that were fed an HFD. Myeloid cell-specific HIF-1α gene deletion protected against HFD-induced inflammation, CLS formation, poor vasculature development in the adipose tissue, and systemic insulin resistance. Despite a reduced expression of Vegfa in epididymal white adipose tissue (eWAT), the preadipocytes and endothelial cells of HIF-1α-deficient mice expressed higher levels of angiogenic factors, including Vegfa, Angpt1, Fgf1, and Fgf10 in accordance with preferable eWAT remodeling. Our in vitro study revealed that lipopolysaccharide-treated bone marrow-derived macrophages directly inhibited the expression of angiogenic factors in 3T3-L1 preadipocytes. Thus, macrophage HIF-1α is involved not only in the formation of CLSs, further enhancing the inflammatory responses, but also in the inhibition of neoangiogenesis in preadipocytes. We concluded that these two pathways contribute to the obesity-related physiology of pathological adipose tissue expansion, thus causing systemic insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Insulin Resistance/genetics , Myeloid Cells/metabolism , 3T3-L1 Cells , Angiopoietin-1/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Female , Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factor 10/metabolism , Glucose Tolerance Test , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Inflammation/etiology , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/metabolism , Vascular Endothelial Growth Factor A/metabolism
14.
Diabetol Int ; 7(1): 59-68, 2016 Mar.
Article in English | MEDLINE | ID: mdl-30603244

ABSTRACT

Chronic inflammation is a pathophysiology of insulin resistance in metabolic diseases, such as obesity and type 2 diabetes. Adipose tissue macrophages (ATMs) play important roles in this inflammatory process. SIRT1 is implicated in the regulation of glucose metabolism in some metabolic tissues, such as liver or skeletal muscle. This study was performed to investigate whether SIRT1 in macrophages played any roles in the regulation of inflammation and glucose metabolism. Myeloid cell-specific SIRT1-knockout mice were originally generated and analyzed under chow-fed and high-fat-fed conditions. Myeloid cell-specific SIRT1 deletion impaired insulin sensitivity and glucose tolerance assessed by the glucose- or insulin-tolerance test, which was associated with the enhanced expression of inflammation-related genes in epididymal adipose tissue of high-fat-fed mice. Interestingly, the M1 ATMs from the SIRT1-knockout mice showed more hypoxic and inflammatory phenotypes than those from control mice. The expressions of some inflammatory genes, such as Il1b and Nos2, which were induced by in vitro hypoxia treatment, were further enhanced by SIRT1 deletion along with the increased acetylation of HIF-1α in cultured macrophages. These results suggest that deletion of SIRT1 in myeloid cells impairs glucose metabolism by enhancing the hypoxia and inflammatory responses in ATMs, thereby possibly representing a novel therapeutic target for metabolic diseases, such as type 2 diabetes.

15.
Diabetol Int ; 7(4): 342-351, 2016 Dec.
Article in English | MEDLINE | ID: mdl-30603285

ABSTRACT

Adipose tissue not only functions as the major energy-storing tissue, but also functions as an endocrine organ that regulates systemic metabolism by releasing various hormones called adipokines. Macrophages play a critical role in maintaining adipocyte health in a lean state and in remodeling during the progression of obesity. Large numbers of classically activated (M1) macrophages accumulate in adipose tissue as adipocytes become larger because of excessive energy conditions, and they adversely affect insulin resistance by triggering local and systemic inflammation. In contrast, alternatively activated (M2) macrophages seem to maintain the health of adipose tissues in a lean state. In addition, they play a role in adapting to excess energy states, because M2 macrophage dysfunction caused by genetic disruption of the M2 gene results in metabolic disorders under high-fat-fed conditions that are probably attributable to their anti-inflammatory functions. Nonetheless, how M2 macrophages contribute to maintaining the health of adipose tissue and therefore to insulin sensitivity is largely unknown. In this article, we review the literature on the role of M1 and M2 macrophages in metabolism, with a special focus on the role of M2 macrophages in adipose tissue. Likewise, we raise topics of M2 macrophages in non-adipose tissues to expand our understanding of macrophage heterogeneity.

16.
Cell Metab ; 21(5): 706-17, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25921090

ABSTRACT

Nicotinamide phosphoribosyltransferase (NAMPT), the key NAD(+) biosynthetic enzyme, has two different forms, intra- and extracellular (iNAMPT and eNAMPT), in mammals. However, the significance of eNAMPT secretion remains unclear. Here we demonstrate that deacetylation of iNAMPT by the mammalian NAD(+)-dependent deacetylase SIRT1 predisposes the protein to secretion in adipocytes. NAMPT mutants reveal that SIRT1 deacetylates lysine 53 (K53) and enhances eNAMPT activity and secretion. Adipose tissue-specific Nampt knockout and knockin (ANKO and ANKI) mice show reciprocal changes in circulating eNAMPT, affecting hypothalamic NAD(+)/SIRT1 signaling and physical activity accordingly. The defect in physical activity observed in ANKO mice is ameliorated by nicotinamide mononucleotide (NMN). Furthermore, administration of a NAMPT-neutralizing antibody decreases hypothalamic NAD(+) production, and treating ex vivo hypothalamic explants with purified eNAMPT enhances NAD(+), SIRT1 activity, and neural activation. Thus, our findings indicate a critical role of adipose tissue as a modulator for the regulation of NAD(+) biosynthesis at a systemic level.


Subject(s)
Adipose Tissue/metabolism , Hypothalamus/metabolism , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Sirtuin 1/metabolism , Acetylation , Adipose Tissue/cytology , Animals , Cell Line , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Nicotinamide Phosphoribosyltransferase/analysis , Nicotinamide Phosphoribosyltransferase/genetics
17.
Endocrinology ; 154(8): 2600-12, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23715867

ABSTRACT

Because oxidative stress promotes insulin resistance in obesity and type 2 diabetes, it is crucial to find effective antioxidant for the purpose of decreasing this threat. In this study, we explored the effect of astaxanthin, a carotenoid antioxidant, on insulin signaling and investigated whether astaxanthin improves cytokine- and free fatty acid-induced insulin resistance in vitro. We examined the effect of astaxanthin on insulin-stimulated glucose transporter 4 (GLUT4) translocation, glucose uptake, and insulin signaling in cultured rat L6 muscle cells using plasma membrane lawn assay, 2-deoxyglucose uptake, and Western blot analysis. Next, we examined the effect of astaxanthin on TNFα- and palmitate-induced insulin resistance. The amount of reactive oxygen species generated by TNFα or palmitate with or without astaxanthin was evaluated by dichlorofluorescein staining. We also compared the effect of astaxanthin on insulin signaling with that of other antioxidants, α-lipoic acid and α-tocopherol. We observed astaxanthin enhanced insulin-stimulated GLUT4 translocation and glucose uptake, which was associated with an increase in insulin receptor substrate-1 tyrosine and Akt phosphorylation and a decrease in c-Jun N-terminal kinase (JNK) and insulin receptor substrate-1 serine 307 phosphorylation. Furthermore, astaxanthin restored TNFα- and palmitate-induced decreases in insulin-stimulated GLUT4 translocation or glucose uptake with a concomitant decrease in reactive oxygen species generation. α-Lipoic acid enhanced Akt phosphorylation and decreased ERK and JNK phosphorylation, whereas α-tocopherol enhanced ERK and JNK phosphorylation but had little effect on Akt phosphorylation. Collectively these findings indicate astaxanthin is a very effective antioxidant for ameliorating insulin resistance by protecting cells from oxidative stress generated by various stimuli including TNFα and palmitate.


Subject(s)
Glucose Transporter Type 4/metabolism , Insulin/pharmacology , Myoblasts/drug effects , Animals , Antioxidants/pharmacology , Blotting, Western , Cell Line , Deoxyglucose/metabolism , Deoxyglucose/pharmacokinetics , Hypoglycemic Agents/pharmacology , Insulin Receptor Substrate Proteins/metabolism , Insulin Resistance/physiology , JNK Mitogen-Activated Protein Kinases/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Phosphorylation/drug effects , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Thioctic Acid/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Xanthophylls/pharmacology , alpha-Tocopherol/pharmacology
18.
Int J Food Microbiol ; 82(3): 203-11, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12593923

ABSTRACT

We examined the acid tolerance and gad mRNA levels of Escherichia coli O157:H7 (three strains) and nonpathogenic E. coli (strains K12, W1485, and B) grown in foods. The E. coli cells (approximately 30,000 cells) were inoculated on the surface of 10 g of solid food samples (asparagus, broccoli, carrot, celery, cucumber, eggplant, ginger, green pepper, onion, potato, radish, tomato and beef) and in 10 ml of cow's milk, cultured statically at 10-25 degrees C for 1-14 days, and subjected to an acid challenge at 37 degrees C for 1 h in LB medium (pH 3.0). When grown at 20 and 25 degrees C in all foods, except for tomato and ginger, the strains showed a stationary-phase specific acid tolerance. The acid tolerance of the O157 strains changed depending on the types of foods (3-10% survival), but was clearly lower than that of the cells grown in EC medium (more than 90% survival). Tomato and ginger induced relatively high acid tolerances (10-30% survival) in the O157 strains irrespective of the growth phase, probably because of their acidity. No remarkable difference was observed in the acid tolerance between the O157 and nonpathogenic strains grown in all foods. When grown at 10 and 15 degrees C in the foods and EC medium, none of the strains showed the stationary-phase specific acid tolerance. In beef, broccoli, celery, potato and radish, the acid tolerance showed a tendency to decrease with the prolonged cultivation time. In other foods, the acid tolerance was almost constant (about 0.1% survival) irrespective of the growth stage. The mRNA level of glutamate decarboxylase genes (gadA and gadB) correlated to the acid tolerance level when the E. coli cells were grown at 25 degrees C, but was very low even in the stationary phase when the E. coli cells were grown at 15 degrees C or below.


Subject(s)
Adaptation, Physiological , Escherichia coli O157/growth & development , Escherichia coli O157/physiology , Escherichia coli Proteins , Food Microbiology , Glutamate Decarboxylase/genetics , Membrane Proteins/genetics , RNA, Messenger/analysis , Vegetables/microbiology , Animals , Escherichia coli/drug effects , Escherichia coli/genetics , Escherichia coli/growth & development , Escherichia coli/physiology , Escherichia coli O157/drug effects , Escherichia coli O157/genetics , Hydrogen-Ion Concentration , Meat/microbiology , Milk/microbiology , Temperature
19.
J Biosci Bioeng ; 94(4): 315-20, 2002.
Article in English | MEDLINE | ID: mdl-16233309

ABSTRACT

We examined the difference between Escherichia coli O157 and non-pathogenic E. coli in their tolerance to spices. Various spices (5 g each) were homogenized at 25 degrees C for 10 min with 5 ml of 70% ethyl alcohol, and the supernatant solutions obtained by centrifugation were used as spice extracts. When the E. coli strains were incubated with each spice extract at concentrations of 0.01% and 0.1%, a noteworthy difference was observed between the O157 and non-pathogenic strains in their tolerance to nutmeg. The populations of the non-pathogenic strains could not be reduced, but those of the O157 strains were remarkably reduced. Antibacterial activity by the nutmeg extract was also found against the enteropathogenic E. coli O111, but not against enterotoxigenic (O6 and O148) and enteroinvasive (O29 and O124) E. coli. When we examined the antibacterial effect of volatile oils in nutmeg on the O157 and non-pathogenic E. coli strains, all O157 strains tested were found to be more sensitive to beta-pinene than non-pathogenic E. coli strains.

SELECTION OF CITATIONS
SEARCH DETAIL
...