101
|
Xu K, Liu S, Zhao X, Zhang X, Fu X, Zhou Y, Xu K, Miao L, Li Z, Li Y, Qiao L, Bao J. Treating hyperuricemia related non-alcoholic fatty liver disease in rats with resveratrol. Biomed Pharmacother 2019; 110:844-849. [DOI: 10.1016/j.biopha.2018.12.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 12/03/2018] [Accepted: 12/07/2018] [Indexed: 02/06/2023] Open
|
102
|
Wen JJ, Gao H, Hu JL, Nie QX, Chen HH, Xiong T, Nie SP, Xie MY. Polysaccharides from fermented Momordica charantia ameliorate obesity in high-fat induced obese rats. Food Funct 2019; 10:448-457. [DOI: 10.1039/c8fo01609g] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Momordica charantia (M. charantia) has been widely used to treat obesity due to its bioactive ingredients.
Collapse
Affiliation(s)
- Jia-Jia Wen
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - He Gao
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Jie-Lun Hu
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Qi-Xing Nie
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Hai-Hong Chen
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Tao Xiong
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Shao-Ping Nie
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| | - Ming-Yong Xie
- State Key Laboratory of Food Science and Technology
- Nanchang University
- Nanchang
- China
| |
Collapse
|
103
|
Xin Y, Wang Y, Chi J, Zhu X, Zhao H, Zhao S, Wang Y. Elevated free fatty acid level is associated with insulin-resistant state in nondiabetic Chinese people. Diabetes Metab Syndr Obes 2019; 12:139-147. [PMID: 30705599 PMCID: PMC6342222 DOI: 10.2147/dmso.s186505] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Free fatty acids (FFAs) are associated with insulin secretion and insulin resistance. However, the associations among FFAs, obesity, and progression from a normal to a prediabetic state are unclear. METHODS Nondiabetic subjects (5,952) were divided in two groups according to their body mass index (BMI): obese subjects (BMI ≥24 kg/m2) and nonobese subjects (BMI <24 kg/m2). Clinical and multiple glucolipid metabolism data were collected. The homeostasis model assessment for insulin resistance (HOMA-IR) and β-cell function (HOMA-β) was used. HbA1c level between 5.7% and 6.4% was considered prediabetic. Nonparametric tests, one-way ANOVA, and linear correlation analysis were performed. R and SPSS 23.0 software programs were used to analyze the results. RESULTS A U-shaped relationship between FFAs and HOMA-IR was observed. After adjusting for potential confounders, the turning points of FFA levels in the curves were 0.54 mmol/L in the nonobese group and 0.61 mmol/L in the obese group. HOMA-IR levels decreased with increasing FFA concentrations before the turning points (regression coefficient [β]= - 0.9, P=0.0111, for the nonobese group; β=0.2, P=0.5094, for the obese group) and then increased (β=0.9, P=0.0069, for the nonobese group; β=1.5, P=0.0263 for the obese group) after the points. Additionally, our study also identified that FFAs were associated with the prediabetes status in obese individuals. CONCLUSION FFA levels were associated with insulin resistance in nondiabetic subjects, and HOMA-IR in nonobese individuals was more sensitive to FFA changes. Monitoring and controlling plasma FFA levels in obese subjects is significant in decreasing insulin resistance and preventing diabetes.
Collapse
Affiliation(s)
- Yanlu Xin
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China, ;
| | - Yunyang Wang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China, ;
| | - Jingwei Chi
- Laboratory of Thyroid Disease, Affiliated Hospital of Qingdao University, Qingdao, China,
| | - Xvhua Zhu
- Department of Endocrinology and Metabolism, Second People's Hospital of Qingdao West Coast, Qingdao, China
| | - Hui Zhao
- Department of Endocrinology and Metabolism, Hiser Medical Center of Qingdao, Qingdao, China
| | - Shihua Zhao
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China, ;
- Laboratory of Thyroid Disease, Affiliated Hospital of Qingdao University, Qingdao, China,
| | - Yangang Wang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Qingdao University, Qingdao, China, ;
| |
Collapse
|
104
|
Heiston EM, Malin SK. Impact of Exercise on Inflammatory Mediators of Metabolic and Vascular Insulin Resistance in Type 2 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1134:271-294. [PMID: 30919343 DOI: 10.1007/978-3-030-12668-1_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of obesity is cornerstone in the etiology of metabolic and vascular insulin resistance and consequently exacerbates glycemic control. Exercise is an efficacious first-line therapy for type 2 diabetes that improves insulin action through, in part, reducing hormone mediated inflammation. Together, improving the coordination of skeletal muscle metabolism with vascular delivery of glucose will be required for optimizing type 2 diabetes and cardiovascular disease treatment.
Collapse
Affiliation(s)
- Emily M Heiston
- Department of Kinesiology, University of Virginia, Charlottesville, VA, USA
| | - Steven K Malin
- Department of Kinesiology, University of Virginia, Charlottesville, VA, USA.
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA.
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
105
|
Yang S, Meng Y, Yan J, Wang N, Xue Z, Zhang H, Fan Y. Polysaccharide-Enriched Fraction from Amillariella Mellea Fruiting Body Improves Insulin Resistance. Molecules 2018; 24:molecules24010046. [PMID: 30583568 PMCID: PMC6337660 DOI: 10.3390/molecules24010046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 12/15/2018] [Accepted: 12/19/2018] [Indexed: 01/14/2023] Open
Abstract
Despite the edible fungus Amillariella mellea possessing a variety of biological activities, its effects on diabetes are still unclear. Polysaccharides are the main bioactive ingredients. In order to destroy the cell wall to obtain more polysaccharides, we used NaOH solution to extract Amillariella mellea fruiting bodies. The alkali extraction (AAMP) was identified as a polysaccharide-enriched fraction. Using type 2 diabetic rats induced by co-treatment of a high fat diet (HFD) and dexamethasone (DEX), we evaluated the hypoglycemic effects of AAMP. The results showed that oral administration of a high dose of AAMP markedly lowered fasting blood glucose, improving glucose intolerance and insulin resistance. AAMP also enhanced the level of LPL and the expressions of two critical lipases ATGL and HSL, leading to a decrease of serum triglyceride. In addition, AAMP specifically suppressed the expression of SREBP-1c, resulting in AAMP observably inhibiting lipid accumulation in the liver. These findings demonstrated that the improvement of AAMP on HFD/DEX-induced insulin resistance was correlated with its regulation of lipid metabolism. Our results indicated that AAMP could be a novel natural drug or health food used for the treatment of diabetes.
Collapse
Affiliation(s)
- Siwen Yang
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Yuhan Meng
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Jingmin Yan
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Na Wang
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Zhujun Xue
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Hang Zhang
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| | - Yuying Fan
- School of Life Sciences, Northeast Normal University, 130024 Jilin, China.
| |
Collapse
|
106
|
Li L, Qi J, Li H. Natural Products Modulating Autophagy Pathway Against the Pathogenesis of Diabetes Mellitus. Curr Drug Targets 2018; 20:96-110. [DOI: 10.2174/1389450119666180726115805] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/07/2018] [Accepted: 06/21/2018] [Indexed: 01/19/2023]
Abstract
Autophagy is a conserved, regulated cellular process for the degradation of abnormal proteins
and disrupted organelles. Literature has described that dysregulation of autophagy is closely related
to the pathogenesis of diabetes mellitus in processes such as impaired pancreatic β cells function,
peripheral insulin resistance and diabetic complications. Emerging evidence indicates that natural
products may possess anti-diabetic activity via regulation of autophagy. In this review, we summarize
natural products targeting the pathogenesis of diabetes mellitus through the regulation of autophagy
and underline possible mechanisms, providing potential drug candidates or therapies for the treatment
of diabetes mellitus.
Collapse
Affiliation(s)
- Linghuan Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jiameng Qi
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hanbing Li
- Institute of Pharmacology, Department of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
107
|
Zhang Y, Wan J, Xu Z, Hua T, Sun Q. Exercise ameliorates insulin resistance via regulating TGFβ-activated kinase 1 (TAK1)-mediated insulin signaling in liver of high-fat diet-induced obese rats. J Cell Physiol 2018; 234:7467-7474. [PMID: 30367484 DOI: 10.1002/jcp.27508] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 09/10/2018] [Indexed: 12/16/2022]
Abstract
Exercise is an effective therapy for insulin resistance. However, the underlying mechanism remains to be elucidated. Previous research demonstrated that TGFβ-activated kinase 1 (TAK1)-dependent signaling plays a crucial character in hepatic insulin resistance. Hepatic ubiquitin specific protease 4 (USP4), USP18, and dual-specificity phosphatases 14 (DUSP14) can suppress TAK1 phosphorylation, besides tumor necrosis factor receptor-associated factor 3 (TRAF3) and tripartite motif 8 (TRIM8) promote its phosphorylation. In this study, we tried to verify our hypothesis that exercise improves insulin resistance in high-fat diet (HFD)-induced obese (DIO) rats via regulating the TAK1 dependent signaling and TAK1 regulators in liver. Forty male Sprague-Dawley rats were randomized into four groups (n = 10): standard diet and sedentary as normal control; fed on HFD and DIO-sedentary; fed on HFD and DIO-chronic exercise; and fed on HFD and DIO-acute exercise. HFD feeding resulted in increased body weight, visceral fat mass, serum FFAs and hepatic lipid deposition, but decreased hepatic glycogen content and insulin sensitivity. Moreover, hepatic TRAF3 and TRIM8 protein levels increased, whereas USP4, USP18, and DUSP14 protein levels were decreased under obese status, which resulted in enhanced TAK1 phosphorylation and impaired insulin signaling. Exercise training, containing chronic and acute mode, both ameliorated insulin resistance. Meanwhile, decreased TAK1, c-Jun N-terminal kinase 1 (JNK1), and insulin receptor substrate 1 (IRS1) phosphorylation enhanced Akt phosphorylation in liver. Moreover, exercise enhanced USP4 and DUSP14 protein levels, whereas decreased TRIM8 protein levels in obese rats' liver. These results showed that exercise triggered a crucial modulation in TAK1-dependent signaling and its regulators in obese rats' liver, and distinct improvement in insulin sensitivity, which provide new insights into the mechanism by which physical exercise improves insulin resistance.
Collapse
Affiliation(s)
- Yong Zhang
- Division of Physiology, Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Jianyong Wan
- Division of Physiology, Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Zhen Xu
- Division of Immunology, The State Key Laboratory of Pharmaceutical Biotechnology, Medical School, Nanjing University, Nanjing, China
| | - Tianmiao Hua
- Division of Neurobiology, Neurobiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Qingyan Sun
- Division of Physiology, Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| |
Collapse
|
108
|
Li JP, Yuan Y, Zhang WY, Jiang Z, Hu TJ, Feng YT, Liu MX. Effect of Radix isatidis polysaccharide on alleviating insulin resistance in type 2 diabetes mellitus cells and rats. J Pharm Pharmacol 2018; 71:220-229. [PMID: 30298631 DOI: 10.1111/jphp.13023] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 09/17/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVES The objective of this paper was to explore the effects of Radix isatidis polysaccharide (RIP) extracted from Radix isatis on alleviating insulin resistance. METHODS The insulin resistance models of 3T3-L1 preadipocytes and type 2 diabetic rats were established to evaluate the insulin resistance activity of RIP. KEY FINDINGS Radix isatidis polysaccharide within the concentration range of 25-100 μg/ml could reduce cell supernatant glucose and TNF-α levels (P < 0.01) and increase the expression of PI-3K P85, Glut4, IRS-1 and Akt protein in symptoms of IR 3T3-L1 preadipocytes. In the meantime, RIP contributed to relieve the weight loss of diabetic rats whose liver weight and liver index were decreased due to the effects of RIP. Experiments in rats also showed that RIP had capacity in reduced serum TC, TG, LDL-C, FFA, FBG, FINS, MDA, ALT, AST activities and increased serum HDL-C, SOD, ISI (P < 0.05 or 0.01). In addition, the oral glucose tolerance in rats was improved (P < 0.05) and liver damage was restored due to RIP. CONCLUSIONS Radix isatidis polysaccharide significantly alleviates insulin resistance in 3T3-L1 preadipocytes and type 2 diabetic rats. These beneficial effects of RIP may associate with their roles in improving the glucose metabolism, lipid metabolism and oxidative stress.
Collapse
Affiliation(s)
- Ji-Ping Li
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, China
| | - Ye Yuan
- Department of Pharmacy & Laboratory, Ya'an Vocational College, Ya'an, China
| | - Wen-You Zhang
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China
| | - Zhen Jiang
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China
| | - Tian-Jiao Hu
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China
| | - Yi-Ting Feng
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China
| | - Mo-Xiang Liu
- Department of Pharmacology, Medical College, Yangzhou University, Yangzhou, China
| |
Collapse
|
109
|
Chung ST, Courville AB, Onuzuruike AU, Galvan-De La Cruz M, Mabundo LS, DuBose CW, Kasturi K, Cai H, Gharib AM, Walter PJ, Garraffo HM, Chacko S, Haymond MW, Sumner AE. Gluconeogenesis and risk for fasting hyperglycemia in Black and White women. JCI Insight 2018; 3:121495. [PMID: 30232289 DOI: 10.1172/jci.insight.121495] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 08/14/2018] [Indexed: 12/24/2022] Open
Abstract
Black women, compared with White women, have high rates of whole-body insulin resistance but a lower prevalence of fasting hyperglycemia and hepatic steatosis. This dissociation of whole-body insulin resistance from fasting hyperglycemia may be explained by racial differences in gluconeogenesis, hepatic fat, or tissue-specific insulin sensitivity. Two groups of premenopausal federally employed women, without diabetes were studied. Using stable isotope tracers, [2H2O] and [6,62-H2]glucose, basal glucose production was partitioned into its components (gluconeogenesis and glycogenolysis) and basal whole-body lipolysis ([2H5]glycerol) was measured. Indices of insulin sensitivity, whole-body (SI), hepatic (HISIGPR), and adipose tissue, were calculated. Hepatic fat was measured by proton magnetic resonance spectroscopy. Black women had less hepatic fat and lower fractional and absolute gluconeogenesis. Whole-body SI, HISIGPR, and adipose tissue sensitivity were similar by race, but at any given level of whole-body SI, Black women had higher HISIGPR. Therefore, fasting hyperglycemia may be a less common early pathological feature of prediabetes in Black women compared with White women, because gluconeogenesis remains lower despite similar whole-body SI.
Collapse
Affiliation(s)
- Stephanie T Chung
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | | | - Anthony U Onuzuruike
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Mirella Galvan-De La Cruz
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Lilian S Mabundo
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Christopher W DuBose
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Kannan Kasturi
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA
| | - Hongyi Cai
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Ahmed M Gharib
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Peter J Walter
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - H Martin Garraffo
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA
| | - Shaji Chacko
- Department of Pediatrics, Children's Nutrition Research Center, US Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, Texas, USA
| | - Morey W Haymond
- Department of Pediatrics, Children's Nutrition Research Center, US Department of Agriculture/Agricultural Research Service, Baylor College of Medicine, Houston, Texas, USA
| | - Anne E Sumner
- National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK), NIH, Bethesda, Maryland, USA.,National Institute of Minority Health and Health Disparities, NIH, Bethesda, Maryland, USA
| |
Collapse
|
110
|
Jung TW, Lee SH, Kim HC, Bang JS, Abd El-Aty AM, Hacımüftüoğlu A, Shin YK, Jeong JH. METRNL attenuates lipid-induced inflammation and insulin resistance via AMPK or PPARδ-dependent pathways in skeletal muscle of mice. Exp Mol Med 2018; 50:1-11. [PMID: 30213948 PMCID: PMC6137187 DOI: 10.1038/s12276-018-0147-5] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 12/25/2022] Open
Abstract
Physical activity has many beneficial effects on metabolic disorders, such as obesity, insulin resistance, and diabetes. Meteorin-like protein (METRNL), a novel secreted protein homologous to the neurotrophin Metrn, is induced after exercise in the skeletal muscle. Herein, we investigated the effects of METRNL on lipid-mediated inflammation and insulin resistance in skeletal muscle via AMP-activated protein kinase (AMPK) or peroxisome proliferator-activated receptor δ (PPARδ). Treatment with METRNL suppressed inflammatory markers, such as nuclear factor κB (NFκB) nuclear translocation, inhibitory κBα (IκBα) phosphorylation, interleukin-6 (IL-6) expression, and pro-inflammatory cytokines (such as TNFα and MCP-1). METRNL treatment also attenuated the impaired insulin response both in palmitate-treated differentiated C2C12 cells and the skeletal muscle of high-fat diet (HFD)-fed mice. Furthermore, METRNL administration rescued glucose intolerance and reduced HFD-induced body weight gain in mice; however, METRNL did not affect calorie intake. METRNL treatment increased AMPK phosphorylation and PPARδ expression both in differentiated C2C12 cells and mouse skeletal muscle. siRNA-mediated suppression of AMPK and PPARδ abrogated the suppressive effects of METRNL on palmitate-induced inflammation and insulin resistance. Moreover, METRNL augmented the mRNA expression of fatty acid oxidation-associated genes, such as carnitine palmitoyltransferase 1 (CPT1), acyl-CoA oxidase (ACO), and fatty acid binding protein 3 (FABP3). siRNAs for AMPK and PPARδ reversed these changes. In the current study, we report for the first time that METRNL alleviates inflammation and insulin resistance and induces fatty acid oxidation through AMPK or PPARδ-dependent signaling in skeletal muscle. Treatment with a hormone that is normally induced following exercise helps alleviate inflammation and insulin resistance in the muscles of obese mice, highlighting a potential new therapeutic strategy for diabetes and other metabolic disorders. Meteorin-like protein (METRNL) is a hormone produced by muscle cells after physical activity and on exposure to cold. A team led by Ji Hoon Jeong and Yong Kyoo Shin from Chung-Ang University
in Seoul, South Korea, observed that levels of METRNL are reduced in mice fed a high-fat diet and in mouse skeletal muscle cells chemically treated to become unresponsive to insulin. Administering METRNL to the cells or mice reduced levels of inflammation, resulting in improved insulin responses. However, these improvements occurred only when certain regulatory molecules were active, revealing a key pathway through which METRNL acts.
Collapse
Affiliation(s)
- Tae Woo Jung
- Research Administration Team, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Sung Hoon Lee
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, Chunchon, Korea
| | - Joon Seok Bang
- College of Pharmacy, Sookmyung Women's University, Seoul, Korea
| | - A M Abd El-Aty
- Department of Pharmacology, Faculty of Veterinary Medicine, Cairo University, 12211, Giza, Egypt.,Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Ahmet Hacımüftüoğlu
- Department of Medical Pharmacology, Medical Faculty, Ataturk University, Erzurum, Turkey
| | - Yong Kyoo Shin
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Korea.
| | - Ji Hoon Jeong
- Department of Pharmacology, College of Medicine, Chung-Ang University, 221, Heuksuk-dong, Dongjak-gu, Seoul, 156-756, Korea.
| |
Collapse
|
111
|
Pflimlin E, Bielohuby M, Korn M, Breitschopf K, Löhn M, Wohlfart P, Konkar A, Podeschwa M, Bärenz F, Pfenninger A, Schwahn U, Opatz T, Reimann M, Petry S, Tennagels N. Acute and Repeated Treatment with 5-PAHSA or 9-PAHSA Isomers Does Not Improve Glucose Control in Mice. Cell Metab 2018; 28:217-227.e13. [PMID: 29937376 DOI: 10.1016/j.cmet.2018.05.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/13/2018] [Accepted: 05/29/2018] [Indexed: 01/08/2023]
Abstract
Fatty acid esters of hydroxylated fatty acids (FAHFAs) were discovered as a novel class of endogenous mammalian lipids whose profound effects on metabolism have been shown. In the current study, in vitro and in vivo the metabolic effects of two of these FAHFAs, namely palmitic acid-5- (or -9) -hydroxy-stearic acid (5- or 9-PAHSA, respectively) were profiled. In DIO mice fed with differentially composed low- or high-fat diets, acute and subchronic treatment with 5-PAHSA and 9-PAHSA alone, or in combination, did not significantly improve the deranged metabolic status. Neither racemic 5- or 9-PAHSA, nor the enantiomers were able to: (1) increase basal or insulin-stimulated glucose uptake in vitro, (2) stimulate GLP-1 release from GLUTag cells, or (3) induce GSIS in rat, mouse, or human islets or in a human pancreatic β cell line. Therefore, our data do not support the further development of PAHSAs or their derivatives for the control of insulin resistance and hyperglycemia.
Collapse
Affiliation(s)
- Elsa Pflimlin
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Maximilian Bielohuby
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany.
| | - Marcus Korn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Kristin Breitschopf
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Matthias Löhn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Paulus Wohlfart
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Anish Konkar
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Michael Podeschwa
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Felix Bärenz
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Anja Pfenninger
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Uwe Schwahn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Till Opatz
- Institut für Organische Chemie, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Marcel Reimann
- Institut für Organische Chemie, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Stefan Petry
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Norbert Tennagels
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany.
| |
Collapse
|
112
|
Diessler S, Jan M, Emmenegger Y, Guex N, Middleton B, Skene DJ, Ibberson M, Burdet F, Götz L, Pagni M, Sankar M, Liechti R, Hor CN, Xenarios I, Franken P. A systems genetics resource and analysis of sleep regulation in the mouse. PLoS Biol 2018; 16:e2005750. [PMID: 30091978 PMCID: PMC6085075 DOI: 10.1371/journal.pbio.2005750] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/06/2018] [Indexed: 12/30/2022] Open
Abstract
Sleep is essential for optimal brain functioning and health, but the biological substrates through which sleep delivers these beneficial effects remain largely unknown. We used a systems genetics approach in the BXD genetic reference population (GRP) of mice and assembled a comprehensive experimental knowledge base comprising a deep "sleep-wake" phenome, central and peripheral transcriptomes, and plasma metabolome data, collected under undisturbed baseline conditions and after sleep deprivation (SD). We present analytical tools to interactively interrogate the database, visualize the molecular networks altered by sleep loss, and prioritize candidate genes. We found that a one-time, short disruption of sleep already extensively reshaped the systems genetics landscape by altering 60%-78% of the transcriptomes and the metabolome, with numerous genetic loci affecting the magnitude and direction of change. Systems genetics integrative analyses drawing on all levels of organization imply α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor trafficking and fatty acid turnover as substrates of the negative effects of insufficient sleep. Our analyses demonstrate that genetic heterogeneity and the effects of insufficient sleep itself on the transcriptome and metabolome are far more widespread than previously reported.
Collapse
Affiliation(s)
- Shanaz Diessler
- Center for Integrative Genomics, University of Lausanne, Switzerland
| | - Maxime Jan
- Center for Integrative Genomics, University of Lausanne, Switzerland
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Yann Emmenegger
- Center for Integrative Genomics, University of Lausanne, Switzerland
| | - Nicolas Guex
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Benita Middleton
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Debra J. Skene
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Mark Ibberson
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Frederic Burdet
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Lou Götz
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Marco Pagni
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Martial Sankar
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Robin Liechti
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Charlotte N. Hor
- Center for Integrative Genomics, University of Lausanne, Switzerland
| | - Ioannis Xenarios
- Center for Integrative Genomics, University of Lausanne, Switzerland
- Vital-IT Systems Biology Division, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Paul Franken
- Center for Integrative Genomics, University of Lausanne, Switzerland
| |
Collapse
|
113
|
Influence of Large-Volume Liposuction on Metabolic and Cardiovascular Health: A Systematic Review. Ann Plast Surg 2018; 79:623-630. [PMID: 28737560 DOI: 10.1097/sap.0000000000001195] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Evidence suggesting that adipose tissue is a metabolically active tissue has generated debate on the effects of large-volume liposuction (LVL) on metabolic and cardiovascular health. Given the inconsistency in the literature, the authors performed a systematic review to identify available evidence in order to elucidate the potential impact of LVL on metabolic markers and cardiovascular risk factors. METHODS A PubMed search using relevant keywords was conducted. Articles were screened using predetermined inclusion and exclusion criteria. Large-volume liposuction was defined as greater than 3.5 L of lipoaspirate. All studies included evaluation of patients' preoperative and postoperative cardiovascular risk factors, inflammatory cytokines, and/or insulin resistance/sensitivity. Relevant studies were evaluated and assigned a level of evidence. RESULTS A total of 12 studies that met the inclusion criteria were reviewed, of which 1 was a continuation of a previous study. All reports were prospective studies, 2 were randomized control trials, and 3 included a control group. A total of 364 patients were pooled for analysis. The mean volume of lipoaspirate was 7440 ± 1934.9 mL. The mean body mass index at baseline and postliposuction was 30.7 and 28.4, respectively. Seven studies reported a trend toward decrease in total cholesterol levels with an overall mean reduction of 0.21 ± 0.05 mmol/L from 4.6 ± 0.79 mmol/L to 4.4 ± 0.74 mmol/L. After LVL, leptin was reported to significantly decrease in 4 studies, and TNF-α was reported to significantly decrease in 2. Adiponectin was reported to significantly increase in 2 studies. IL-6 decreased significantly in 2 studies. Two studies included participants with type II diabetes mellitus, whereas 10 studies evaluated insulin sensitivity. Of these, 6 studies reported improvement in insulin sensitivity. Six studies represented level IV and 6 represented level II evidence. CONCLUSIONS Liposuction is among the most common aesthetic procedures performed with advances that make it possible to remove considerable amount of adipose tissue within a short period. Current data, although conflicting, appear to support the notion that LVL can affect cardiovascular risk factors, metabolic balance, and insulin resistance in positive ways. Future research with prospective studies is needed to clarify the role of LVL in improving overall health.
Collapse
|
114
|
Hong S, Song W, Zushin PJH, Liu B, Jedrychowski MP, Mina AI, Deng Z, Cabarkapa D, Hall JA, Palmer CJ, Aliakbarian H, Szpyt J, Gygi SP, Tavakkoli A, Lynch L, Perrimon N, Banks AS. Phosphorylation of Beta-3 adrenergic receptor at serine 247 by ERK MAP kinase drives lipolysis in obese adipocytes. Mol Metab 2018; 12:25-38. [PMID: 29661693 PMCID: PMC6001906 DOI: 10.1016/j.molmet.2018.03.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/21/2018] [Accepted: 03/24/2018] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The inappropriate release of free fatty acids from obese adipose tissue stores has detrimental effects on metabolism, but key molecular mechanisms controlling FFA release from adipocytes remain undefined. Although obesity promotes systemic inflammation, we find activation of the inflammation-associated Mitogen Activated Protein kinase ERK occurs specifically in adipose tissues of obese mice, and provide evidence that adipocyte ERK activation may explain exaggerated adipose tissue lipolysis observed in obesity. METHODS AND RESULTS We provide genetic and pharmacological evidence that inhibition of the MEK/ERK pathway in human adipose tissue, mice, and flies all effectively limit adipocyte lipolysis. In complementary findings, we show that genetic and obesity-mediated activation of ERK enhances lipolysis, whereas adipose tissue specific knock-out of ERK2, the exclusive ERK1/2 protein in adipocytes, dramatically impairs lipolysis in explanted mouse adipose tissue. In addition, acute inhibition of MEK/ERK signaling also decreases lipolysis in adipose tissue and improves insulin sensitivity in obese mice. Mice with decreased rates of adipose tissue lipolysis in vivo caused by either MEK or ATGL pharmacological inhibition were unable to liberate sufficient White Adipose Tissue (WAT) energy stores to fuel thermogenesis from brown fat during a cold temperature challenge. To identify a molecular mechanism controlling these actions, we performed unbiased phosphoproteomic analysis of obese adipose tissue at different time points following acute pharmacological MEK/ERK inhibition. MEK/ERK inhibition decreased levels of adrenergic signaling and caused de-phosphorylation of the β3-adrenergic receptor (β3AR) on serine 247. To define the functional implications of this phosphorylation, we showed that CRISPR/Cas9 engineered cells expressing wild type β3AR exhibited β3AR phosphorylation by ERK2 and enhanced lipolysis, but this was not seen when serine 247 of β3AR was mutated to alanine. CONCLUSION Taken together, these data suggest that ERK activation in adipocytes and subsequent phosphorylation of the β3AR on S247 are critical regulatory steps in the enhanced adipocyte lipolysis of obesity.
Collapse
Affiliation(s)
- Shangyu Hong
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Song
- Department of Genetics, Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Peter-James H Zushin
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Bingyang Liu
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | | | - Amir I Mina
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Zhaoming Deng
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Dimitrije Cabarkapa
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Jessica A Hall
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Colin J Palmer
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Hassan Aliakbarian
- Department of Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, 02115, USA
| | - John Szpyt
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Ali Tavakkoli
- Department of Surgery, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, 02115, USA
| | - Lydia Lynch
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Norbert Perrimon
- Department of Genetics, Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, 02115, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
115
|
Rotimi SO, Rotimi OA, Adelani IB, Onuzulu C, Obi P, Okungbaye R. Stevioside modulates oxidative damage in the liver and kidney of high fat/low streptozocin diabetic rats. Heliyon 2018; 4:e00640. [PMID: 29872771 PMCID: PMC5986550 DOI: 10.1016/j.heliyon.2018.e00640] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 05/08/2018] [Accepted: 05/25/2018] [Indexed: 12/29/2022] Open
Abstract
This study investigated the potential of stevioside to prevent oxidative DNA damage in the liver and kidney of type 2 diabetes mellitus (T2DM) using high fat-low streptozocin rat model. Rats were treated daily with 12.5, 25 and 50 mg/kg stevioside orally for 21 days. Levels of biomarkers of T2DM, lipid profile and oxidative stress were assayed spectrophotometrically. The DNA ladder assay method was used to assess DNA fragmentation in the liver and kidney while computational analysis was used to predict the mechanisms of antidiabetic properties of stevioside. Stevioside significantly (p < 0.05) decreased the levels of plasma glucose, insulin, dipeptidyl peptidase IV and activities of kidney angiotensin converting enzyme. Stevioside significantly reduced oxidative stress by decreasing the levels of lipid peroxidation and nitric oxide in the liver and kidney; thereby, reducing the extent of DNA fragmentation in the liver and kidney of the diabetic rats. The in silico analysis showed that the ability of stevioside to exert these effects is linked to its inhibition of beta-adrenergic receptor kinase and G-protein-coupled receptor kinase. The results of this study suggest that the prevention of DNA fragmentation may be an additional benefit of the use of stevioside in the management of T2DM.
Collapse
|
116
|
Zhang Y, Wan J, Liu S, Hua T, Sun Q. Exercise induced improvements in insulin sensitivity are concurrent with reduced NFE2/miR-432-5p and increased FAM3A. Life Sci 2018; 207:23-29. [PMID: 29802941 DOI: 10.1016/j.lfs.2018.05.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/14/2018] [Accepted: 05/22/2018] [Indexed: 10/16/2022]
Abstract
AIMS Little is known regarding whether the NFE2/miR-423-5p and FAM3A-ATP-Akt pathway in liver mediates exercise allured alleviation of insulin resistance connected with diet-induced obesity. This research inquired the influence of exercise on liver insulin sensitivity and whole body insulin resistance in high-fat diet fed rats. MATERIALS AND METHODS Forty male Sprague-Dawley rats at seven-week-old were assigned to four groups at random: standard diet as normal control group (NC, n = 10), high-fat diet group (HFD, n = 10), high-fat diet with chronic exercise intervention group (HFD-CE, n = 10) and high-fat diet with acute exercise intervention group (HFD-AE, n = 10). KEY FINDINGS Compared with rats fed with a standard diet, eight-week high-fat diet feeding lead to elevated body weight, visceral fat content and serum FFAs, and decreased insulin sensitivity index. Moreover, high-fat diet enhanced NFE2 protein expression and miR-423-5p level, decreased FAM3A mRNA and protein expression, ATP level and Akt phosphorylation in liver. In contrast, physical exercise, both chronic and acute exercise alleviated whole body insulin resistance, reduced hepatic NFE2 and miR-423-5p expression, and serum FFAs level, meanwhile enhanced FAM3A mRNA and protein expression, ATP level and Akt phosphorylation in liver. The current findings indicated that exercise in diet-induced obesity, both chronic and acute, induce a momentous regulation in NFE2/miR-423-5p and FAM3A-ATP-Akt pathway in liver, and improve hepatic insulin sensitivity and whole body insulin resistance. SIGNIFICANCE All these results supply crucial evidence in our comprehending of the molecular mechanism that connected exercise to an alleviation of insulin resistance.
Collapse
Affiliation(s)
- Yong Zhang
- Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Jianyong Wan
- Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Shiqiang Liu
- Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Tianmiao Hua
- Neurobiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China
| | - Qingyan Sun
- Physiology Laboratory of College of Life Sciences, Anhui Normal University, Wuhu, China.
| |
Collapse
|
117
|
Morris JK, Uy RAZ, Vidoni ED, Wilkins HM, Archer AE, Thyfault JP, Miles JM, Burns JM. Effect of APOE ε4 Genotype on Metabolic Biomarkers in Aging and Alzheimer's Disease. J Alzheimers Dis 2018; 58:1129-1135. [PMID: 28550261 DOI: 10.3233/jad-170148] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Alzheimer's disease (AD) may have heterogeneous pathophysiological underpinnings, with risk factors including apolipoprotein rmvarep4 (APOE4) genotype and insulin resistance. We hypothesized that distinct phenotypes exist within AD. We examined APOE4 and metabolic biomarkers in 338 subjects (n = 213 nondemented (ND), n = 125 AD). We further characterized steady state free fatty acid (FFA) levels in a subset of 45 participants who had also participated in a hyperinsulinemic-euglycemic clamp. Insulin resistance (HOMA-IR) was elevated in AD versus ND (p = 0.04) and in APOE4 noncarriers versus carriers (p < 0.01). This was driven by increased fasting insulin in AD versus ND (p < 0.01) and in APOE4 non-carriers versus carriers (p = 0.01). Fasting glucose was not different. In subjects who underwent a clamp, there was a group x genotype interaction on FFA levels during hyperinsulinemia (p = 0.03). APOE4 non-carriers with AD had higher FFA levels, while APOE4 carriers with AD exhibited lower FFA levels. Metabolic dysfunction is overrepresented in individuals with AD dementia who do not carry the APOE4 allele. This suggests that important subsets of AD phenotypes may exist that diverge metabolically.
Collapse
Affiliation(s)
- Jill K Morris
- Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Roxanne Adeline Z Uy
- Department of Endocrinology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Eric D Vidoni
- Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Heather M Wilkins
- Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Ashley E Archer
- Department of Molecular and Integrative Physiology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - John M Miles
- Department of Endocrinology, University of Kansas School of Medicine, Kansas City, KS, USA
| | - Jeffrey M Burns
- Department of Neurology, University of Kansas School of Medicine, Kansas City, KS, USA
| |
Collapse
|
118
|
Fiamoncini J, Rundle M, Gibbons H, Thomas EL, Geillinger-Kästle K, Bunzel D, Trezzi JP, Kiselova-Kaneva Y, Wopereis S, Wahrheit J, Kulling SE, Hiller K, Sonntag D, Ivanova D, van Ommen B, Frost G, Brennan L, Bell J, Daniel H. Plasma metabolome analysis identifies distinct human metabotypes in the postprandial state with different susceptibility to weight loss-mediated metabolic improvements. FASEB J 2018; 32:5447-5458. [PMID: 29718708 DOI: 10.1096/fj.201800330r] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Health has been defined as the capability of the organism to adapt to challenges. In this study, we tested to what extent comprehensively phenotyped individuals reveal differences in metabolic responses to a standardized mixed meal tolerance test (MMTT) and how these responses change when individuals experience moderate weight loss. Metabolome analysis was used in 70 healthy individuals. with profiling of ∼300 plasma metabolites during an MMTT over 8 h. Multivariate analysis of plasma markers of fatty acid catabolism identified 2 distinct metabotype clusters (A and B). Individuals from metabotype B showed slower glucose clearance, had increased intra-abdominal adipose tissue mass and higher hepatic lipid levels when compared with individuals from metabotype A. An NMR-based urine analysis revealed that these individuals also to have a less healthy dietary pattern. After a weight loss of ∼5.6 kg over 12 wk, only the subjects from metabotype B showed positive changes in the glycemic response during the MMTT and in markers of metabolic diseases. Our study in healthy individuals demonstrates that more comprehensive phenotyping can reveal discrete metabotypes with different outcomes in a dietary intervention and that markers of lipid catabolism in plasma could allow early detection of the metabolic syndrome.-Fiamoncini, J., Rundle, M., Gibbons, H., Thomas, E. L., Geillinger-Kästle, K., Bunzel, D., Trezzi, J.-P., Kiselova-Kaneva, Y., Wopereis, S., Wahrheit, J., Kulling, S. E., Hiller, K., Sonntag, D., Ivanova, D., van Ommen, B., Frost, G., Brennan, L., Bell, J. Daniel, H. Plasma metabolome analysis identifies distinct human metabotypes in the postprandial state with different susceptibility to weight loss-mediated metabolic improvements.
Collapse
Affiliation(s)
- Jarlei Fiamoncini
- Department of Food and Nutrition, Technische Universität München, Freising-Weihenstephan, Germany
| | - Milena Rundle
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
| | - Helena Gibbons
- University College Dublin (UCD) School of Agriculture and Food Science, Institute of Food and Health, Dublin, Ireland
| | - E Louise Thomas
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | | | - Diana Bunzel
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner Institut, Karlsruhe, Germany
| | - Jean-Pierre Trezzi
- Integrated Biobank of Luxembourg, Dudelange, Luxembourg.,Centre for Systems Biomedicine, Esch-sur-Alzette, Luxembourg
| | - Yoana Kiselova-Kaneva
- Department of Biochemistry, Molecular Medicine, and Nutrigenomics, Medical University-Varna, Varna, Bulgaria
| | - Suzan Wopereis
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | | | - Sabine E Kulling
- Department of Safety and Quality of Fruit and Vegetables, Max Rubner Institut, Karlsruhe, Germany
| | - Karsten Hiller
- Braunschweig Integrated Centre of Systems Biology, University of Braunschweig, Braunschweig, Germany.,Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Denise Sonntag
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Diana Ivanova
- Department of Biochemistry, Molecular Medicine, and Nutrigenomics, Medical University-Varna, Varna, Bulgaria
| | - Ben van Ommen
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research (TNO), Zeist, The Netherlands
| | - Gary Frost
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Imperial College London, London, United Kingdom
| | - Lorraine Brennan
- University College Dublin (UCD) School of Agriculture and Food Science, Institute of Food and Health, Dublin, Ireland
| | - Jimmy Bell
- Research Centre for Optimal Health, Department of Life Sciences, University of Westminster, London, United Kingdom
| | - Hannelore Daniel
- Department of Food and Nutrition, Technische Universität München, Freising-Weihenstephan, Germany
| |
Collapse
|
119
|
Han Q, Cao Y, Gathaiya N, Kemp BJ, Jensen MD. Free fatty acid flux measured using [1- 11C]palmitate positron emission tomography and [U- 13C]palmitate in humans. Am J Physiol Endocrinol Metab 2018; 314:E413-E417. [PMID: 29046281 PMCID: PMC6008058 DOI: 10.1152/ajpendo.00284.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
PET radiopharmaceuticals can noninvasively measure free fatty acid (FFA) tissue uptake. Investigators often use PET scan-derived data to calculate FFA flux. We tested whether the [1-11C]palmitate PET measures of palmitate flux provide results equivalent to a continuous infusion of [U-13C]palmitate. Nine volunteers participated in study 1 to evaluate whether a rapidly (10-20 s) given bolus of [1-11C]palmitate affects calculated flux results. Thirty volunteers participated in study 2, which was identical to study 1 except that the [1-11C]palmitate bolus was given over 1 min. Volunteers in both studies also received a continuous intravenous infusion of [U-13C]palmitate. Plasma palmitate concentrations and enrichment were measured by liquid chromatography-mass spectrometry. The PET/CT images were analyzed on a workstation running PMOD. Palmitate flux was estimated using PET time-activity curve (TAC) data from regions of interest in the left ventricle (LV) and aorta both with and without hybrid TACs that employed the 11CO2-corrected data for the first 5 min and the 11CO2-corrected blood radioactivity for the remainder of the PET scan. Palmitate flux in study 1 measured with PET [1-11C]palmitate and [U-13C]palmitate were not correlated, and the PET [1-11C]palmitate flux was significantly less than the [U-13C]palmitate measured flux. In study 2, the palmitate flux using PET [1-11C]palmitate hybrid LV models provided closer mean estimates of [U-13C]palmitate measured flux. The best PET calculation approaches predicted 64% of the interindividual variance in [U-13C]palmitate measured flux. Palmitate kinetics measured using [1-11C]palmitate/PET do not provide the same palmitate kinetic results as the continuous infusion [U-13C]palmitate approach.
Collapse
Affiliation(s)
- Qiaojun Han
- Endocrine Research Unit, Mayo Clinic , Rochester, Minnesota
| | - Yanli Cao
- Endocrine Research Unit, Mayo Clinic , Rochester, Minnesota
| | | | - Bradley J Kemp
- Division of Medical Physics, Department of Radiology, Mayo Clinic , Rochester, Minnesota
| | | |
Collapse
|
120
|
Batchuluun B, Al Rijjal D, Prentice KJ, Eversley JA, Burdett E, Mohan H, Bhattacharjee A, Gunderson EP, Liu Y, Wheeler MB. Elevated Medium-Chain Acylcarnitines Are Associated With Gestational Diabetes Mellitus and Early Progression to Type 2 Diabetes and Induce Pancreatic β-Cell Dysfunction. Diabetes 2018; 67:885-897. [PMID: 29436377 PMCID: PMC5910003 DOI: 10.2337/db17-1150] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 01/30/2018] [Indexed: 12/30/2022]
Abstract
Specific circulating metabolites have emerged as important risk factors for the development of diabetes. The acylcarnitines (acylCs) are a family of metabolites known to be elevated in type 2 diabetes (T2D) and linked to peripheral insulin resistance. However, the effect of acylCs on pancreatic β-cell function is not well understood. Here, we profiled circulating acylCs in two diabetes cohorts: 1) women with gestational diabetes mellitus (GDM) and 2) women with recent GDM who later developed impaired glucose tolerance (IGT), new-onset T2D, or returned to normoglycemia within a 2-year follow-up period. We observed a specific elevation in serum medium-chain (M)-acylCs, particularly hexanoyl- and octanoylcarnitine, among women with GDM and individuals with T2D without alteration in long-chain acylCs. Mice treated with M-acylCs exhibited glucose intolerance, attributed to impaired insulin secretion. Murine and human islets exposed to elevated levels of M-acylCs developed defects in glucose-stimulated insulin secretion and this was directly linked to reduced mitochondrial respiratory capacity and subsequent ability to couple glucose metabolism to insulin secretion. In conclusion, our study reveals that an elevation in circulating M-acylCs is associated with GDM and early stages of T2D onset and that this elevation directly impairs β-cell function.
Collapse
Affiliation(s)
| | - Dana Al Rijjal
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kacey J Prentice
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Judith A Eversley
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Elena Burdett
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Haneesha Mohan
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Erica P Gunderson
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Ying Liu
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
121
|
Inhibition of insulin resistance by PGE1 via autophagy-dependent FGF21 pathway in diabetic nephropathy. Sci Rep 2018; 8:9. [PMID: 29311680 PMCID: PMC5758726 DOI: 10.1038/s41598-017-18427-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/11/2017] [Indexed: 12/22/2022] Open
Abstract
Insulin resistance is a critical process in the initiation and progression of diabetic nephropathy (DN). Alprostadil (Prostaglandin E1, PGE1) had protective effects on renal function. However, it is unknown whether PGE1 inhibited insulin resistance in renal tubule epithelial cells via autophagy, which plays a protective role in DN against insulin resistance. Insulin resistance was induced by palmitic acid (PA) in human HK-2 cells, shown as the decrease of insulin-stimulated AKT phosphorylation, glucose transporter-4 (GLUT4), glucose uptake and enhanced phosphorylation of insulin receptor substrate 1(IRS-1) at site serine 307 (pIRS-1ser307) and downregulated expression of IRS-1. Along with less abundance of p62, autophagy markers LC3B and Beclin-1 significantly increased in HK-2 cells exposed to PA. Such abnormal changes were significantly reversed by PGE1, which mimicked the role of autophagy gene 7 small interfering RNA (ATG7 siRNA). Furthermore, PGE1 promoted the protein expression of autophagy-related fibroblast growth factor-21 (FGF21), which alleviated insulin resistance. Results from western blotting and immunohistochemistry indicated that PGE1 remarkably restored autophagy, insulin resistance and the FGF21 expression in rat kidney of type 2 diabetes mellitus (T2DM). Collectively, we demonstrated the potential protection of PGE1 on insulin resistance in renal tubules via autophagy-dependent FGF21 pathway in preventing the progression of DN.
Collapse
|
122
|
Hou W, Meng X, Zhao A, Zhao W, Pan J, Tang J, Huang Y, Li H, Jia W, Liu F, Jia W. Development of Multimarker Diagnostic Models from Metabolomics Analysis for Gestational Diabetes Mellitus (GDM). Mol Cell Proteomics 2017; 17:431-441. [PMID: 29282297 DOI: 10.1074/mcp.ra117.000121] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 12/06/2017] [Indexed: 12/12/2022] Open
Abstract
Although metabolomics are desirable to understand the pathophysiology of gestational diabetes mellitus (GDM), comprehensive metabolomic studies of GDM are rare. We aimed to offer a holistic view of metabolites alteration in GDM patients and investigate the possible multimarker models for GDM diagnosis. Biochemical parameters and perinatal data of 131 GDM cases and 138 controls were collected. Fasting serum samples at 75 g oral glucose tolerance test were used for metabolites by ultra performance liquid chromatography-quadrupole-time of flight-mass spectrometry, ultra performance liquid chromatography-triple triple-quadrupole-mass spectrometry and gas chromatography- time-of- flight mass spectrometry platforms. Significant changes were observed in free fatty acids, bile acids, branched chain amino acids, organic acids, lipids and organooxygen compounds between two groups. In receiver operating characteristic (ROC) analysis, different combinations of candidate biomarkers and metabolites in multimarker models achieved satisfactory discriminative abilities for GDM, with the values of area under the curve (AUC) ranging from 0.721 to 0.751. Model consisting of body mass index (BMI), retinol binding protein 4 (RBP4), n-acetylaspartic acid and C16:1 (cis-7) manifested the best discrimination [AUC 0.751 (95% CI: 0.693-0.809), p < 0.001], followed by model consisting of BMI, Cystatin C, acetylaspartic acid and 6,7-diketoLCA [AUC 0.749 (95% CI: 0.691-0.808), p < 0.001]. Metabolites alteration reflected disorders of glucose metabolism, lipid metabolism, amino acid metabolism, bile acid metabolism as well as intestinal flora metabolism in GDM state. Multivariate models combining clinical markers and metabolites have the potential to differentiate GDM subjects from healthy controls.
Collapse
Affiliation(s)
- Wolin Hou
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China
| | - Xiyan Meng
- §Department of Obstetrics and Gynecology, Shanghai Clinical Center for Severe Maternal Rescue, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Aihua Zhao
- ¶Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Weijing Zhao
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China
| | - Jiemin Pan
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China
| | - Junling Tang
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China
| | - Yajuan Huang
- §Department of Obstetrics and Gynecology, Shanghai Clinical Center for Severe Maternal Rescue, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Huaping Li
- §Department of Obstetrics and Gynecology, Shanghai Clinical Center for Severe Maternal Rescue, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wei Jia
- ¶Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Fang Liu
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China;
| | - Weiping Jia
- From the ‡Shanghai Key Laboratory of Diabetes, Department of Endocrinology & Metabolism, Shanghai Jiao-Tong University Affiliated Sixth People's Hospital, Shanghai Clinical Medical Center of Diabetes, Shanghai Key Clinical Center of Metabolic Diseases, Shanghai Institute for Diabetes, Shanghai, China
| |
Collapse
|
123
|
Feist PE, Loughran EA, Stack MS, Hummon AB. Quantitative proteomic analysis of murine white adipose tissue for peritoneal cancer metastasis. Anal Bioanal Chem 2017; 410:1583-1594. [PMID: 29282499 DOI: 10.1007/s00216-017-0813-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 11/30/2017] [Accepted: 12/06/2017] [Indexed: 12/15/2022]
Abstract
Cancer metastasis risk increases in older individuals, but the mechanisms for this risk increase are unclear. Many peritoneal cancers, including ovarian cancer, preferentially metastasize to peritoneal fat depots. However, there is a dearth of studies exploring aged peritoneal adipose tissue in the context of cancer. Because adipose tissue produces signals which influence several diseases including cancer, proteomics of adipose tissue in aged and young mice may provide insight into metastatic mechanisms. We analyzed mesenteric, omental, and uterine adipose tissue groups from the peritoneal cavities of young and aged C57BL/6J mouse cohorts with a low-fraction SDS-PAGE gelLC-MS/MS method. We identified 2308 protein groups and quantified 2167 groups, among which several protein groups showed twofold or greater abundance differences between the aged and young cohorts. Cancer-related gene products previously identified as significant in another age-related study were found altered in this study. Several gene products known to suppress proliferation and cellular invasion were found downregulated in the aged cohort, including R-Ras, Arid1a, and heat shock protein β1. In addition, multiple protein groups were identified within single cohorts, including the proteins Cd11a, Stat3, and Ptk2b. These data suggest that adipose tissue is a strong candidate for analysis to identify possible contributors to cancer metastasis in older subjects. The results of this study, the first of its kind using uterine adipose tissue, contribute to the understanding of the role of adipose tissue in age-related alteration of oncogenic pathways, which may help elucidate the mechanisms of increased metastatic tumor burden in the aged. Graphical abstract We analyzed mesenteric, omental, and uterine adipose tissue groups from the peritoneal cavities of young and aged C57BL/6J mouse cohorts with a low-fraction SDS-PAGE gelLC-MS/MS method. These fat depots are preferential sites for many peritoneal cancers. The results of this study, the first of its kind using uterine adipose tissue, contribute to the understanding of the role of adipose tissue in age-related alteration of oncogenic pathways, which may help elucidate the mechanisms of increased metastatic tumor burden in the aged.
Collapse
Affiliation(s)
- Peter E Feist
- Integrated Biomedical Sciences Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, 251 140B McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Elizabeth A Loughran
- Integrated Biomedical Sciences Program, University of Notre Dame, Notre Dame, IN, 46556, USA
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, 251 140B McCourtney Hall, Notre Dame, IN, 46556, USA
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, 251 140B McCourtney Hall, Notre Dame, IN, 46556, USA
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, 251 140B McCourtney Hall, Notre Dame, IN, 46556, USA.
| |
Collapse
|
124
|
Hetta MH, Owis AI, Haddad PS, Eid HM. The fatty acid-rich fraction of Eruca sativa (rocket salad) leaf extract exerts antidiabetic effects in cultured skeletal muscle, adipocytes and liver cells. PHARMACEUTICAL BIOLOGY 2017; 55:810-818. [PMID: 28112007 PMCID: PMC6130626 DOI: 10.1080/13880209.2017.1280687] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 11/04/2016] [Accepted: 01/06/2017] [Indexed: 05/27/2023]
Abstract
CONTEXT Eruca sativa Mill. (Brassicaceae), commonly known as rocket salad, is a popular leafy-green vegetable with many health benefits. OBJECTIVE To evaluate the antidiabetic activities of this plant in major insulin-responsive tissues. MATERIALS AND METHODS Five E. sativa leaf extracts of varying polarity were prepared (aqueous extract, 70% and 95% ethanol extracts, the n-hexane-soluble fraction of the 95% ethanol extract (ES3) and the defatted 95% ethanol extract). Eruca sativa extracts were investigated through a variety of cell-based in vitro bioassays for antidiabetic activities in C2C12 skeletal muscle cells, H4IIE hepatocytes and 3T3-L1 adipocytes. Guided by the results of these bioassays, ES3 was fractionated into the saponifiable (SM) and the unspaonifiable (USM) fractions. Glucose uptake was measured using [3H]-deoxy-glucose, while the effects on hepatic glucose-6-phosphatase (G6Pase) and adipogenesis were assessed using Wako AutoKit Glucose and AdipoRed assays, respectively. RESULTS ES3 and its SM fraction significantly stimulated glucose uptake with EC50 values of 8.0 and 5.8 μg/mL, respectively. Both extracts significantly inhibited G6Pase activity (IC50 values of 4.8 and 9.3 μg/mL, respectively). Moreover, ES3 and SM showed significant adipogenic activities with EC50 of 4.3 and 6.1 μg/mL, respectively. Fatty acid content of SM was identified by GC-MS. trans-Vaccenic and palmitoleic acids were the major unsaturated fatty acids, while palmitic and azelaic acids were the main saturated fatty acids. DISCUSSION AND CONCLUSION These findings indicate that ES3 and its fatty acid-rich fraction exhibit antidiabetic activities in insulin-responsive cell lines and may hence prove useful for the treatment of type 2 diabetes.
Collapse
Affiliation(s)
- Mona H. Hetta
- Department of Pharmacognosy, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Asmaa I. Owis
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Pierre S. Haddad
- Department of Pharmacology and Physiology, Natural Health Products and Metabolic Diseases Laboratory, Université de Montréal, Montréal, Quebec, Canada
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines and Montreal Diabetes Research Center, Montréal, Quebec, Canada
| | - Hoda M. Eid
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
- Department of Pharmacology and Physiology, Natural Health Products and Metabolic Diseases Laboratory, Université de Montréal, Montréal, Quebec, Canada
- Canadian Institutes of Health Research Team in Aboriginal Antidiabetic Medicines and Montreal Diabetes Research Center, Montréal, Quebec, Canada
| |
Collapse
|
125
|
Differential acute and chronic responses in insulin action in cultured myotubes following from nondiabetic severely obese humans following gastric bypass surgery. Surg Obes Relat Dis 2017; 13:1853-1862. [DOI: 10.1016/j.soard.2017.05.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 05/16/2017] [Accepted: 05/17/2017] [Indexed: 01/26/2023]
|
126
|
Takato T, Iwata K, Murakami C, Wada Y, Sakane F. Chronic administration of myristic acid improves hyperglycaemia in the Nagoya-Shibata-Yasuda mouse model of congenital type 2 diabetes. Diabetologia 2017; 60:2076-2083. [PMID: 28707095 DOI: 10.1007/s00125-017-4366-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/06/2017] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Previously, we demonstrated that myristic acid (14:0) increases levels of diacylglycerol kinase (DGK) δ, a key enzyme involved in type 2 diabetes exacerbation, and enhances glucose uptake in C2C12 myotube cells. Moreover, results from a population-based cohort study suggest that consumption of high-fat dairy products, which contain high amounts of myristic acid, is associated with a lower risk of developing type 2 diabetes. Taken together, we hypothesised that intake of myristic acid reduces type 2 diabetes risk in vivo. The aim of this study was to examine the glucose-lowering effect of myristic acid in Nagoya-Shibata-Yasuda (NSY) mice, a spontaneous model for studying obesity-related type 2 diabetes. METHODS Male NSY mice were orally administered vehicle (n = 9), 300 mg/kg of myristic acid (n = 14) or 300 mg/kg of palmitic acid (16:0) (n = 9) every other day from 4 weeks of age. Glucose and insulin tolerance tests were performed at weeks 18, 24 and 30, and weeks 20 and 26, respectively. DGKδ levels were measured in skeletal muscle from 32-36-week-old NSY mice via western blot. RESULTS Chronic oral administration of myristic acid ameliorated glucose tolerance (24-28% decrease in blood glucose levels during glucose tolerance tests) and reduced insulin-responsive blood glucose levels (~20% decrease) in male NSY mice compared with vehicle and palmitic acid groups at 24-30 weeks of age (the age at which the severity of type 2 diabetes is exacerbated in NSY mice). Myristic acid also attenuated the increase in body weight seen in NSY mice. Furthermore, the fatty acid increased DGKδ levels (~1.6-fold) in skeletal muscle of NSY mice. CONCLUSIONS/INTERPRETATION These results suggest that the chronic oral administration of myristic acid improves hyperglycaemia by decreasing insulin-responsive glucose levels and reducing body weight, and that the fatty acid accounts for the diabetes protective properties of high-fat dairy products. Myristic acid is a potential candidate for the prevention and treatment of type 2 diabetes mellitus and its related diseases.
Collapse
Affiliation(s)
- Tamae Takato
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Kai Iwata
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Yuko Wada
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, 1-33 Yayoi-cho, Inage-ku, Chiba, 263-8522, Japan.
| |
Collapse
|
127
|
Varani K, Vincenzi F, Merighi S, Gessi S, Borea PA. Biochemical and Pharmacological Role of A1 Adenosine Receptors and Their Modulation as Novel Therapeutic Strategy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1051:193-232. [DOI: 10.1007/5584_2017_61] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
128
|
San-Millán I, Brooks GA. Assessment of Metabolic Flexibility by Means of Measuring Blood Lactate, Fat, and Carbohydrate Oxidation Responses to Exercise in Professional Endurance Athletes and Less-Fit Individuals. Sports Med 2017. [DOI: 10.1007/s40279-017-0751-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
129
|
Kim M, Song K, Kim YS. Alantolactone improves palmitate-induced glucose intolerance and inflammation in both lean and obese states in vitro: Adipocyte and adipocyte-macrophage co-culture system. Int Immunopharmacol 2017; 49:187-194. [PMID: 28599253 DOI: 10.1016/j.intimp.2017.05.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 05/29/2017] [Accepted: 05/31/2017] [Indexed: 11/26/2022]
Abstract
Obesity is characterized by a massive infiltration of the adipose tissue by macrophages. Adipocytes, together with macrophages create a crosstalk between inflammation and insulin resistance. Excess saturated FFA, such as palmitate, absorbed via the portal system may cause glucose intolerance and inflammation, which leads to insulin resistance. In this study, we aimed to evaluate the potency of alantolactone (AL), a sesquiterpene lactone isolated from Inula helenium in reducing palmitate-induced glucose intolerance, fat accumulation, and inflammation in 3T3-L1 adipocytes and adipocyte-macrophage co-culture system (3T3-L1-RAW264.7). We observed that palmitate reduced glucose uptake and increased fat accumulation, which indicated dysfunctional adipocytes with inadequate lipid storage. However, AL treatment reversed these changes in a dose-dependent manner (P<0.05). Palmitate activated c-Jun N-terminal kinases (JNK) and IκB kinase β/α (IKKβ/α) phosphorylation, and increased the levels of the proinflammatory cytokines (tumor necrosis factor-α and interleukin-6 [IL-6]) and chemokines (monocyte chemoattractant protein-1 [MCP-1]). AL treatment selectively reduced JNK-associated mitogen-activated protein kinase pathway (JNK and extracellular signal-regulated kinase phosphorylation). However, it did not affect NF-κB pathway in adipocytes. In addition, AL decreased the gene expression of JNK upregulating factor, toll-like receptor-4 (TLR4), suggesting inhibition of TLR4-JNK signaling. Moreover, it reduced inflammation-associated IL-6 and MCP-1 mRNA levels in both adipocytes and adipocyte-macrophage system. Our study showed that palmitate treatment led to adipocyte dysfunction and macrophage infiltration; however, AL improved palmitate-induced glucose intolerance and inflammation. These findings suggest that AL may inhibit obesity-induced insulin resistance and improve glucose homeostasis and inflammation in insulin target tissues.
Collapse
Affiliation(s)
- Minjee Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Kwangho Song
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
130
|
Estimation of insulin secretion, glucose uptake by tissues, and liver handling of glucose using a mathematical model of glucose-insulin homeostasis in lean and obese mice. Heliyon 2017. [PMID: 28626803 PMCID: PMC5463011 DOI: 10.1016/j.heliyon.2017.e00310] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Destruction of the insulin-producing β-cells is the key determinant of diabetes mellitus regardless of their types. Due to their anatomical location within the islets of Langerhans scattered throughout the pancreas, it is difficult to monitor β-cell function and mass clinically. To this end, we propose to use a mathematical model of glucose-insulin homeostasis to estimate insulin secretion, glucose uptake by tissues, and hepatic handling of glucose. We applied the mathematical model by Lombarte et al. (2013) to compare various rate constants representing glucose-insulin homeostasis between lean (11% fat)- and high fat diet (HFD; 45% fat)-fed mice. Mice fed HFD (n = 12) for 3 months showed significantly higher body weights (49.97 ± 0.52 g vs. 29.86 ± 0.46 g), fasting blood glucose levels (213.08 ± 10.35 mg/dl vs. 121.91 ± 2.26 mg/dl), and glucose intolerance compared to mice fed lean diet (n = 12). Mice were injected with 1 g/kg glucose intraperitoneally and blood glucose levels were measured at various intervals for 120 min. We performed simulation using Arena™ software based on the mathematical model and estimated the rate constants (9 parameters) for various terms in the differential equations using OptQuest™. The simulated data fit accurately to the observed data for both lean and obese mice, validating the use of the mathematical model in mice at different stages of diabetes progression. Among 9 parameters, 5 parameters including basal insulin, k2 (rate constant for insulin-dependent glucose uptake to tissues), k3 (rate constant for insulin-independent glucose uptake to tissues), k4 (rate constant for liver glucose transfer), and Ipi (rate constant for insulin concentration where liver switches from glucose release to uptake) were significantly different between lean- and HFD-fed mice. Basal blood insulin levels, k3, and Ipi were significantly elevated but k2 and k4 were reduced in mice fed a HFD compared to those fed a lean diet. Non-invasive assessment of the key components of glucose-insulin homeostasis including insulin secretion, glucose uptake by tissues, and hepatic handling of glucose may be helpful for individualized drug therapy and designing a customized control algorithm for the artificial pancreas.
Collapse
|
131
|
Thondam SK, Daousi C, Wilding JPH, Holst JJ, Ameen GI, Yang C, Whitmore C, Mora S, Cuthbertson DJ. Glucose-dependent insulinotropic polypeptide promotes lipid deposition in subcutaneous adipocytes in obese type 2 diabetes patients: a maladaptive response. Am J Physiol Endocrinol Metab 2017; 312:E224-E233. [PMID: 28073779 DOI: 10.1152/ajpendo.00347.2016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Revised: 11/28/2016] [Accepted: 01/02/2017] [Indexed: 12/19/2022]
Abstract
Glucose-dependent insulinotropic polypeptide (GIP) beyond its insulinotropic effects may regulate postprandial lipid metabolism. Whereas the insulinotropic action of GIP is known to be impaired in type 2 diabetes mellitus (T2DM), its adipogenic effect is unknown. We hypothesized that GIP is anabolic in human subcutaneous adipose tissue (SAT) promoting triacylglycerol (TAG) deposition through reesterification of nonesterified fatty acids (NEFA), and this effect may differ according to obesity status or glucose tolerance. Twenty-three subjects categorized into four groups, normoglycemic lean (n = 6), normoglycemic obese (n = 6), obese with impaired glucose regulation (IGR; n = 6), and obese T2DM (n = 5), participated in a double-blind, randomized, crossover study involving a hyperglycemic clamp with a 240-min GIP infusion (2 pmol·kg-1·min-1) or normal saline. Insulin, NEFA, SAT-TAG content, and gene expression of key lipogenic enzymes were determined before and immediately after GIP/saline infusions. GIP lowered NEFA concentrations in the obese T2DM group despite diminished insulinotropic activity (mean NEFA AUC0-4 h ± SE, 41,992 ± 9,843 µmol·l-1·min-1 vs. 71,468 ± 13,605 with placebo, P = 0.039, 95% CI: 0.31-0.95). Additionally, GIP increased SAT-TAG in obese T2DM (1.78 ± 0.4 vs 0.86 ± 0.1-fold with placebo, P = 0.043, 95% CI: 0.1-1.8). Such effect with GIP was not observed in other three groups despite greater insulinotropic activity. Reduction in NEFA concentration with GIP correlated with adipose tissue insulin resistance for all subjects (Pearson, r = 0.56, P = 0.005). There were no significant gene expression changes in key SAT lipid metabolism enzymes. In conclusion, GIP appears to promote fat accretion and thus may exacerbate obesity and insulin resistance in T2DM.
Collapse
Affiliation(s)
- Sravan K Thondam
- Obesity and Endocrinology Research Group, University Hospital Aintree, Liverpool, United Kingdom
| | - Christina Daousi
- Obesity and Endocrinology Research Group, University Hospital Aintree, Liverpool, United Kingdom
| | - John P H Wilding
- Obesity and Endocrinology Research Group, University Hospital Aintree, Liverpool, United Kingdom
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| | - Jens J Holst
- NovoNordisk Foundation Center for Basic Metabolic Research and Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Gulizar I Ameen
- Department of Cellular and Molecular Physiology University of Liverpool, Liverpool, United Kingdom; and
| | - Chenjing Yang
- Department of Cellular and Molecular Physiology University of Liverpool, Liverpool, United Kingdom; and
| | - Catherine Whitmore
- Obesity and Endocrinology Research Group, University Hospital Aintree, Liverpool, United Kingdom
| | - Silvia Mora
- Department of Cellular and Molecular Physiology University of Liverpool, Liverpool, United Kingdom; and
| | - Daniel J Cuthbertson
- Obesity and Endocrinology Research Group, University Hospital Aintree, Liverpool, United Kingdom
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
132
|
Chen W, Balland E, Cowley MA. Hypothalamic Insulin Resistance in Obesity: Effects on Glucose Homeostasis. Neuroendocrinology 2017; 104:364-381. [PMID: 28122381 DOI: 10.1159/000455865] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 01/04/2017] [Indexed: 01/05/2023]
Abstract
The central link between obesity and type 2 diabetes is the development of insulin resistance. To date, it is still not clear whether hyperinsulinemia causes insulin resistance, which underlies the pathogenesis of obesity-associated type 2 diabetes, owing to the sophisticated regulatory mechanisms that exist in the periphery and in the brain. In recent years, accumulating evidence has demonstrated the existence of insulin resistance within the hypothalamus. In this review, we have integrated the recent discoveries surrounding both central and peripheral insulin resistance to provide a comprehensive overview of insulin resistance in obesity and the regulation of systemic glucose homeostasis. In particular, this review will discuss how hyperinsulinemia and hyperleptinemia in obesity impair insulin sensitivity in tissues such as the liver, skeletal muscle, adipose tissue, and the brain. In addition, this review highlights insulin transport into the brain, signaling pathways associated with hypothalamic insulin receptor expression in the regulation of hepatic glucose production, and finally the perturbation of systemic glucose homeostasis as a consequence of central insulin resistance. We also suggest future approaches to overcome both central and peripheral insulin resistance to treat obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Weiyi Chen
- Department of Physiology/Biomedical Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | | |
Collapse
|
133
|
Pereira MJ, Skrtic S, Katsogiannos P, Abrahamsson N, Sidibeh CO, Dahgam S, Månsson M, Risérus U, Kullberg J, Eriksson JW. Impaired adipose tissue lipid storage, but not altered lipolysis, contributes to elevated levels of NEFA in type 2 diabetes. Degree of hyperglycemia and adiposity are important factors. Metabolism 2016; 65:1768-1780. [PMID: 27832864 DOI: 10.1016/j.metabol.2016.09.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 09/06/2016] [Accepted: 09/22/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Elevated levels of circulating non-esterified fatty acids (NEFA) mediate many adverse metabolic effects. In this work we aim to determine the impact of type 2 diabetes (T2D), glycemic control and obesity on lipolysis regulation. DESIGN AND PARTICIPANTS 20 control and 20 metformin-treated T2D subjects were matched for sex (10M/10 F), age (58±11 vs 58±9 y) and BMI (30.8±4.6 vs 30.7±4.9kg/m2). In vivo lipolysis was assessed during a 3h-OGTT with plasma glycerol and NEFA levels. Subcutaneous adipose tissue (SAT) biopsies were obtained to measure mRNA and metabolite levels of factors related to lipolysis and lipid storage and to assess in vitro lipolysis in isolated subcutaneous adipocytes. RESULTS Plasma NEFA AUC during the OGTT where higher 30% (P=0.005) in T2D than in control subjects, but plasma glycerol AUC and subcutaneous adipocyte lipolysis in vitro were similar, suggesting that adipose tissue lipolysis is not altered. Expression in SAT of genes involved in lipid storage (FABP4, DGAT1, FASN) were reduced in T2D subjects compared with controls, but no differences were seen for genes involved in lipolysis. T2D subjects had elevated markers of beta-oxidation, α-hydroxybutyrate (1.4-fold, P<0.01) and β-hydroxybutyrate (1.7-fold, P<0.05) in plasma. In multivariate analysis, HbA1c, visceral adipose tissue volume and sex (male) were significantly associated with NEFA AUC in T2D subjects. CONCLUSIONS In T2D subjects, NEFA turnover is impaired, but not due to defects in lipolysis or lipid beta-oxidation. Impaired adipose NEFA re-esterification or de novo lipogenesis is likely to contribute to higher NEFA plasma levels in T2D. The data suggest that hyperglycemia and adiposity are important contributing factors for the regulation of plasma NEFA concentrations.
Collapse
Affiliation(s)
- Maria J Pereira
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Stanko Skrtic
- AstraZeneca R&D, Mölndal, Sweden; Department of Endocrinology, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | | | - Cherno O Sidibeh
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | | | | | - Ulf Risérus
- Clinical Nutrition and Metabolism, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Joel Kullberg
- Radiology, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jan W Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
134
|
Nemati R, Lu J, Tura A, Smith G, Murphy R. Acute Changes in Non-esterified Fatty Acids in Patients with Type 2 Diabetes Receiving Bariatric Surgery. Obes Surg 2016; 27:649-656. [PMID: 27530911 DOI: 10.1007/s11695-016-2323-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND The purpose of this study was to compare acute changes of non-esterified fatty acids (NEFA) in relation to beta cell function (BCF) and insulin resistance in obese patients with type 2 diabetes (T2D) who underwent laparoscopic gastric bypass (GBP), laparoscopic sleeve gastrectomy (SG) or very low calorie diet (VLCD). METHODS In a non-randomised study, fasting plasma samples were collected from 38 obese patients with T2D, matched for age, body mass index (BMI) and glycaemic control, who underwent GBP (11) or SG (14) or VLCD (13). Samples were collected the day before and 3 days after the intervention, during a 75-g oral glucose tolerance test. Glucose, insulin, c-peptide, glucagon like peptide-1 (GLP-1) and gastric inhibitory polypeptide (GIP) were measured, and individual NEFAs were measured using a triple-quadrupole liquid chromatography-mass spectrometry (LC-MS/MS). BCF by mathematical modelling and insulin resistance were estimated. RESULTS Palmitic acid significantly decreased after each intervention. Monounsaturated/polyunsaturated ratio (MUFA/PUFA) and unsaturated/saturated fat ratios increased after each intervention. BCF was improved only after VLCD. Linoleic acid was positively correlated with total insulin secretion (p = 0.03). Glucose sensitivity correlated with palmitic acid (p = 0.01), unsaturated/saturated ratio (p = 0.0008) and MUFA/PUFA (p = 0.009). HOMA-IR correlated with stearic acid (p = 0.03), unsaturated/saturated ratio (p = 0.005) and MUFA/PUFA (p = 0.009). GIP AUC0-120 correlated with stearic acid (p = 0.04), but not GLP-1. CONCLUSIONS GBP, SG and VLCD have similar acute effects on decreasing palmitic acid. Several NEFAs correlated with BCF parameters and HOMA-IR.
Collapse
Affiliation(s)
- Reza Nemati
- School of Applied Sciences, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand
| | - Jun Lu
- School of Applied Sciences, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.,School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, Auckland University of Technology, Auckland, New Zealand.,Institute of Biomedical Technology, Auckland University of Technology, Auckland, New Zealand
| | - Andrea Tura
- Metabolic Unit, Institute of Neuroscience, National Research Council, 35127, Padua, Italy
| | - Greg Smith
- Department of Pharmacology, University of New South Wales, Sydney, Australia
| | - Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand.
| |
Collapse
|
135
|
Pharmacological Evaluation of Naproxen Metal Complexes on Antinociceptive, Anxiolytic, CNS Depressant, and Hypoglycemic Properties. Adv Pharmacol Sci 2016; 2016:3040724. [PMID: 27478435 PMCID: PMC4958467 DOI: 10.1155/2016/3040724] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/17/2016] [Indexed: 11/22/2022] Open
Abstract
Purpose. The present study was designed to investigate the antinociceptive, anxiolytic, CNS depressant, and hypoglycemic effects of the naproxen metal complexes. Methods. The antinociceptive activity was evaluated by acetic acid-induced writhing method and radiant heat tail-flick method while anxiolytic activity was evaluated by elevated plus maze model. The CNS depressant activity of naproxen metal complexes was assessed using phenobarbitone-induced sleeping time test and the hypoglycemic test was performed using oral glucose tolerance test. Results. Metal complexes significantly (P < 0.001) reduced the number of abdominal muscle contractions induced by 0.7% acetic acid solution in a dose dependent manner. At the dose of 25 mg/kg body weight p.o. copper, cobalt, and zinc complexes exhibited higher antinociceptive activity having 59.15%, 60.56%, and 57.75% of writhing inhibition, respectively, than the parent ligand naproxen (54.93%). In tail-flick test, at both doses of 25 and 50 mg/kg, the copper, cobalt, silver, and zinc complexes showed higher antinociceptive activity after 90 minutes than the parent drug naproxen. In elevated plus maze (EPM) model the cobalt and zinc complexes of naproxen showed significant anxiolytic effects in dose dependent manner, while the copper, cobalt, and zinc complexes showed significant CNS depressant and hypoglycemic activity. Conclusion. The present study demonstrated that copper, cobalt, and zinc complexes possess higher antinociceptive, anxiolytic, CNS depressant, and hypoglycemic properties than the parent ligand.
Collapse
|
136
|
Hershkop K, Besor O, Santoro N, Pierpont B, Caprio S, Weiss R. Adipose Insulin Resistance in Obese Adolescents Across the Spectrum of Glucose Tolerance. J Clin Endocrinol Metab 2016; 101:2423-31. [PMID: 27054297 PMCID: PMC4891802 DOI: 10.1210/jc.2016-1376] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
CONTEXT Adipocytes represent an important insulin-responsive tissue taking an active part in glucose metabolism. OBJECTIVE This study sought to assess adipose tissue insulin resistance (IR) across the spectrum of glucose tolerance and to test its relation with free fatty acid (FFA) suppression during an oral glucose tolerance test (OGTT). DESIGN AND SETTING A cross-sectional analysis of a pediatric clinic-derived cohort of obese adolescents. PATIENTS OR OTHER PARTICIPANTS Participants age 7-20 y with a body mass index that exceeded the 95th percentile for their age and sex. INTERVENTION(S) A standard oral glucose tolerance test. MAIN OUTCOME MEASURES The adipose tissue insulin resistance index (calculated as the product of fasting insulin and FFA concentrations) (Adipose IR) and the area under curve of FFAs during the OGTT were compared between glucose tolerance categories. RESULTS A total of 962 obese children and adolescents participated in this study. Adipose IR significantly increased across glucose tolerance categories (P for trend < .001). Within the normal glucose tolerance participants, an increase in adipose IR was observed related to an increase in 2-hr glucose levels. In a subsample of participants who underwent abdominal imaging for determination of lipid partitioning (n = 115), a tight relation of visceral fat (r = 0.34; P < .001) and the visceral/sc fat ratio (r = 0.55; P < .001) with the Adipose IR index was evident. Greater area under the curve FFAs (lower FFA suppression) during the OGTT was evident with worsening glucose tolerance (P for trend < .001). Glucose tolerance category, degree of obesity (body mass index-z score), IL-6, and low adiponectin emerged as significant predictors of the Adipose IR. CONCLUSIONS Adipose IR is associated with reduced suppression of FFAs during the OGTT and with an altered adipocytokine profile. The negative relation with insulin secretion deserves further longitudinal investigation in the context of deteriorating glucose tolerance.
Collapse
Affiliation(s)
- Karen Hershkop
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| | - Omri Besor
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| | - Nicola Santoro
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| | - Bridget Pierpont
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| | - Sonia Caprio
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| | - Ram Weiss
- Department of Human Metabolism and Nutrition (K.H., O.B., R.W.), Braun School of Public Health, Hebrew University, Jerusalem 91120, Israel; and the Department of Pediatrics (N.S., B.P., S.C.), Yale School of Medicine, New Haven, Connecticut 06510
| |
Collapse
|
137
|
Yu J, Zheng J, Liu XF, Feng ZL, Zhang XP, Cao LL, Zhou ZP. Exercise improved lipid metabolism and insulin sensitivity in rats fed a high-fat diet by regulating glucose transporter 4 (GLUT4) and musclin expression. ACTA ACUST UNITED AC 2016; 49:e5129. [PMID: 27143172 PMCID: PMC4855995 DOI: 10.1590/1414-431x20165129] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 02/04/2016] [Indexed: 12/19/2022]
Abstract
This study aimed to evaluate the effects of exercise training on triglyceride deposition and the expression of musclin and glucose transporter 4 (GLUT4) in a rat model of insulin resistance. Thirty male Sprague-Dawley rats (8 weeks old, weight 160±10 g) were fed a high-fat diet (40% calories from fat) and randomly divided into high-fat control group and swimming intervention group. Rats fed with standard food served as normal control. We found that 8-week swimming intervention significantly decreased body weight (from 516.23±46.27 to 455.43±32.55 g) and visceral fat content (from 39.36±2.50 to 33.02±2.24 g) but increased insulin sensitivity index of the rats fed with a high-fat diet. Moreover, swimming intervention improved serum levels of TG (from 1.40±0.83 to 0.58±0.26 mmol/L) and free fatty acids (from 837.80±164.25 to 556.38±144.77 μEq/L) as well as muscle triglycerides deposition (from 0.55±0.06 to 0.45±0.02 mmol/g) in rats fed a high-fat diet. Compared with rats fed a standard food, musclin expression was significantly elevated, while GLUT4 expression was decreased in the muscles of rats fed a high-fat diet. In sharp contrast, swimming intervention significantly reduced the expression of musclin and increased the expression of GLUT4 in the muscles of rats fed a high-fat diet. In conclusion, increased musclin expression may be associated with insulin resistance in skeletal muscle, and exercise training improves lipid metabolism and insulin sensitivity probably by upregulating GLUT4 and downregulating musclin.
Collapse
Affiliation(s)
- J Yu
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - J Zheng
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - X F Liu
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - Z L Feng
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - X P Zhang
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - L L Cao
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| | - Z P Zhou
- The First Hospital of Jiujiang City, Jiangxi Province, China, Department of Endocrinology, Jiujiang Affiliated Hospital of Nanchang University, Jiujiang, Jiangxi Province, China
| |
Collapse
|
138
|
Ozaki KI, Awazu M, Tamiya M, Iwasaki Y, Harada A, Kugisaki S, Tanimura S, Kohno M. Targeting the ERK signaling pathway as a potential treatment for insulin resistance and type 2 diabetes. Am J Physiol Endocrinol Metab 2016; 310:E643-E651. [PMID: 26860984 DOI: 10.1152/ajpendo.00445.2015] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 02/04/2016] [Indexed: 12/21/2022]
Abstract
Extracellular signal-regulated kinase (ERK) has been implicated in the development of insulin resistance associated with obesity and type 2 diabetes mellitus. We have now examined the potential of pharmacological targeting of the ERK pathway with MEK (ERK kinase) inhibitors (PD184352 and PD0325901) for the treatment of obesity-associated insulin resistance. The effects of PD184352 and PD0325901 on the expression of adipocytokines and lipolysis activity were thus examined in 3T3-L1 adipocytes maintained in long-term culture as a model of adipocyte hypertrophy. Leptin receptor-deficient (db/db) mice and high-fat diet-fed KKAy mice, both of which are models of type 2 diabetes, were also treated orally with PD184352 to examine its effects on the diabetic condition. ERK activity was increased in hypertrophic 3T3-L1 adipocytes as well as in adipose tissue of db/db mice and high-fat diet-fed KKAy mice, and this enhanced ERK signaling was associated with dysregulation of adipocytokine expression and increased lipolysis activity. Specific blockade of the ERK pathway in hypertrophic 3T3-L1 adipocytes by MEK inhibitors ameliorated the dysregulation of adipocytokine expression and suppressed the enhanced lipolysis activity. Furthermore, repeated oral administration of PD184352 normalized hyperglycemia and hyperlipidemia and improved insulin sensitivity and glucose tolerance in the diabetic mice. These results suggest that sustained activation of the ERK pathway in adipocytes is associated with the pathogenesis of type 2 diabetes and that selective blockade of this pathway with MEK inhibitors warrants further study as a promising approach to the treatment of insulin resistance and type 2 diabetes.
Collapse
Affiliation(s)
- Kei-Ichi Ozaki
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Midori Awazu
- Department of Pediatrics, Keio University School of Medicine, Tokyo, Japan
| | - Mayuko Tamiya
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Yuka Iwasaki
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Aya Harada
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Satomi Kugisaki
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Susumu Tanimura
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| | - Michiaki Kohno
- Laboratory of Cell Regulation, Department of Pharmaceutical Sciences, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Japan; and
| |
Collapse
|
139
|
Rojas-Morales P, Tapia E, Pedraza-Chaverri J. β-Hydroxybutyrate: A signaling metabolite in starvation response? Cell Signal 2016; 28:917-23. [PMID: 27083590 DOI: 10.1016/j.cellsig.2016.04.005] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/09/2016] [Indexed: 02/08/2023]
Abstract
Ketone bodies β-hydroxybutyrate (BHB) and acetoacetate are important metabolic substrates for energy production during prolonged fasting. However, BHB also has signaling functions. Through several metabolic pathways or processes, BHB modulates nutrient utilization and energy expenditure. These findings suggest that BHB is not solely a metabolic intermediate, but also acts as a signal to regulate metabolism and maintain energy homeostasis during nutrient deprivation. We briefly summarize the metabolism and emerging physiological functions of ketone bodies and highlight the potential role for BHB as a signaling molecule in starvation response.
Collapse
Affiliation(s)
- Pedro Rojas-Morales
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edilia Tapia
- Laboratorio de Fisiopatología Renal, Departamento de Nefrología, Instituto Nacional de Cardiología - Ignacio Chávez, Mexico City, Mexico
| | - José Pedraza-Chaverri
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
140
|
Combined Non-alcoholic Fatty Liver Disease and Type 2 Diabetes Mellitus: Sleeve Gastrectomy or Gastric Bypass?—a Controlled Matched Pair Study of 34 Patients. Obes Surg 2015; 26:1867-74. [DOI: 10.1007/s11695-015-2006-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
141
|
Broussard JL, Kolka CM, Castro AVB, Asare Bediako I, Paszkiewicz RL, Szczepaniak EW, Szczepaniak LS, Knutson KL, Kim SP, Bergman RN. Elevated nocturnal NEFA are an early signal for hyperinsulinaemic compensation during diet-induced insulin resistance in dogs. Diabetologia 2015; 58:2663-70. [PMID: 26254577 PMCID: PMC4591216 DOI: 10.1007/s00125-015-3721-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Accepted: 07/14/2015] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS A normal consequence of increased energy intake and insulin resistance is compensatory hyperinsulinaemia through increased insulin secretion and/or reduced insulin clearance. Failure of compensatory mechanisms plays a central role in the pathogenesis of type 2 diabetes mellitus; consequently, it is critical to identify in vivo signal(s) involved in hyperinsulinaemic compensation. We have previously reported that high-fat feeding leads to an increase in nocturnal NEFA concentration. We therefore designed this study to test the hypothesis that elevated nocturnal NEFA are an early signal for hyperinsulinaemic compensation for insulin resistance. METHODS Blood sampling was conducted in male dogs to determine 24 h profiles of NEFA at baseline and during high-fat feeding with and without acute nocturnal NEFA suppression using a partial A1 adenosine receptor agonist. RESULTS High-fat feeding increased nocturnal NEFA and reduced insulin sensitivity, effects countered by an increase in acute insulin response to glucose (AIR(g)). Pharmacological NEFA inhibition after 8 weeks of high-fat feeding lowered NEFA to baseline levels and reduced AIR(g) with no effect on insulin sensitivity. A significant relationship emerged between nocturnal NEFA levels and AIR(g). This relationship indicates that the hyperinsulinaemic compensation induced in response to high-fat feeding was prevented when the nocturnal NEFA pattern was returned to baseline. CONCLUSIONS/INTERPRETATION Elevated nocturnal NEFA are an important signal for hyperinsulinaemic compensation during diet-induced insulin resistance.
Collapse
Affiliation(s)
- Josiane L Broussard
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA.
| | - Cathryn M Kolka
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Ana V B Castro
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Isaac Asare Bediako
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Rebecca L Paszkiewicz
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Edward W Szczepaniak
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Lidia S Szczepaniak
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | | | - Stella P Kim
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Richard N Bergman
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, Los Angeles, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| |
Collapse
|
142
|
Ni Y, Zhao L, Yu H, Ma X, Bao Y, Rajani C, Loo LW, Shvetsov YB, Yu H, Chen T, Zhang Y, Wang C, Hu C, Su M, Xie G, Zhao A, Jia W, Jia W. Circulating Unsaturated Fatty Acids Delineate the Metabolic Status of Obese Individuals. EBioMedicine 2015; 2:1513-1522. [PMID: 26629547 PMCID: PMC4634820 DOI: 10.1016/j.ebiom.2015.09.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 09/03/2015] [Accepted: 09/03/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Obesity is not a homogeneous condition across individuals since about 25-40% of obese individuals can maintain healthy status with no apparent signs of metabolic complications. The simple anthropometric measure of body mass index does not always reflect the biological effects of excessive body fat on health, thus additional molecular characterizations of obese phenotypes are needed to assess the risk of developing subsequent metabolic conditions at an individual level. METHODS To better understand the associations of free fatty acids (FFAs) with metabolic phenotypes of obesity, we applied a targeted metabolomics approach to measure 40 serum FFAs from 452 individuals who participated in four independent studies, using an ultra-performance liquid chromatograph coupled to a Xevo G2 quadruple time-of-flight mass spectrometer. FINDINGS FFA levels were significantly elevated in overweight/obese subjects with diabetes compared to their healthy counterparts. We identified a group of unsaturated fatty acids (UFAs) that are closely correlated with metabolic status in two groups of obese individuals who underwent weight loss intervention and can predict the recurrence of diabetes at two years after metabolic surgery. Two UFAs, dihomo-gamma-linolenic acid and palmitoleic acid, were also able to predict the future development of metabolic syndrome (MS) in a group of obese subjects. INTERPRETATION These findings underscore the potential role of UFAs in the MS pathogenesis and also as important markers in predicting the risk of developing diabetes in obese individuals or diabetes remission after a metabolic surgery.
Collapse
Key Words
- AA, arachidonic acid
- BMI, body mass index
- CVD, cardiovascular disease
- DAG, diacylglycerol
- DBP, diastolic blood pressure
- DGLA, dihomo-gamma-linolenic acid
- DNL, de novo lipogenesis
- FATPs, fatty acid transport proteins
- FFA, free fatty acids
- Free fatty acids
- GLA, γ-linolenic acid
- HA, heptadecanoic acid
- HDL, high-density lipoprotein
- HO, metabolically healthy obese
- HbA1c, glycated hemoglobin
- Insulin resistance
- LA, linoleic acid
- LDL, low-density lipoprotein
- MS, metabolic syndrome
- MUFA, monounsaturated acid
- Metabolic syndrome
- NAFLD, nonalcoholic fatty liver disease
- NW, normal weight
- OGTT, oral glucose tolerance test
- OPLS-DA, orthogonal partial least square discriminant analysis
- Obesity
- PA, palmitoleic acid
- PUFA, polyunsaturated fatty acid
- RSD, relative standard deviation
- SBP, systolic blood pressure
- SCD, stearoyl-CoA desaturase
- SFA, saturated fatty acid
- SHDS, the Shanghai Diabetes Study
- SHOS, the Shanghai Obesity Study
- T2D, type 2 diabetes
- TC, total cholesterol
- TG, triglycerides
- Type 2 diabetes
- UFA, unsaturated fatty acid
- UO, metabolically unhealthy obese
- Unsaturated fatty acids
- VLCD, very low carbohydrate diet
Collapse
Affiliation(s)
- Yan Ni
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Linjing Zhao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Haoyong Yu
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| | - Xiaojing Ma
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| | - Yuqian Bao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| | - Cynthia Rajani
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | | | | | - Herbert Yu
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Tianlu Chen
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Yinan Zhang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Congrong Wang
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| | - Cheng Hu
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| | - Mingming Su
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Guoxiang Xie
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Aihua Zhao
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Wei Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| | - Weiping Jia
- Shanghai Key Laboratory of Diabetes Mellitus and Center for Translational Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
- Department of Endocrinology and Metabolism, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai 200233, China
| |
Collapse
|
143
|
Treviño S, Sánchez-Lara E, Sarmiento-Ortega VE, Sánchez-Lombardo I, Flores-Hernández JÁ, Pérez-Benítez A, Brambila-Colombres E, González-Vergara E. Hypoglycemic, lipid-lowering and metabolic regulation activities of metforminium decavanadate (H2Metf)3 [V10O28]·8H2O using hypercaloric-induced carbohydrate and lipid deregulation in Wistar rats as biological model. J Inorg Biochem 2015; 147:85-92. [PMID: 25920353 DOI: 10.1016/j.jinorgbio.2015.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Revised: 04/01/2015] [Accepted: 04/01/2015] [Indexed: 12/14/2022]
Abstract
Because of the increasing global spread of type 2 diabetes mellitus, there is a need to develop new antidiabetic agents. Recently we have synthesized new decavanadates using metformin as counterion. In particular, the compound containing three metforminium dications has been obtained in high yield and has been completely characterized. Biological studies using Wistar rats that have been fed with a high caloric diet inducing insulin resistance and metabolic syndrome were carried out. Results of the impact on key biochemical parameters mediated by metformin alone and the new compound are here presented. The metforminium decavanadate (H2Metf)3[V10O28]·8H2O, abbreviated as Metf-V10O28, was shown to have pharmacological potential as a hypoglycemic, lipid-lowering and metabolic regulator, since the resulting compound made of the two components with antidiabetic activities, reduces both dosage and time of administration (twice a week). Hence, due to the beneficial effects induced by the metforminium decavanadate we recommend to continue the exploration into the mechanism and toxicology of this new compound.
Collapse
Affiliation(s)
- Samuel Treviño
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Eduardo Sánchez-Lara
- Centro de Química, ICUAP, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Víctor Enrique Sarmiento-Ortega
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Irma Sánchez-Lombardo
- Centro de Química, ICUAP, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - José Ángel Flores-Hernández
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Aarón Pérez-Benítez
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Eduardo Brambila-Colombres
- Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico
| | - Enrique González-Vergara
- Centro de Química, ICUAP, Benemérita Universidad Autónoma de Puebla, 14 Sur y Av. San Claudio, Col. San Manuel, CP 72570 Puebla, PUE, Mexico.
| |
Collapse
|
144
|
Xin W, Zhao X, Liu L, Xu Y, Li Z, Chen L, Wang X, Yi F, Wan Q. Acetyl-CoA carboxylase 2 suppression rescues human proximal tubular cells from palmitic acid induced lipotoxicity via autophagy. Biochem Biophys Res Commun 2015; 463:364-9. [PMID: 26022126 DOI: 10.1016/j.bbrc.2015.05.070] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 05/18/2015] [Indexed: 02/02/2023]
Abstract
Autophagy is a catabolic process that degrades damaged proteins and organelles in mammalian cells. Although acetyl-CoA carboxylase 2 (ACC2) plays a crucial role in the fatty acid metabolism, it keeps unknown whether ACC2 is associated with autophagic activity. The present work was designed to investigate the effects of ACC2 on palmitic acid (PA) induced lipotoxicity in human proximal tubular cells and the putative role of autophagy in this process. Here we show that autophagy was induced by PA in HK-2 cells. Moreover, the PA induced autophagy was regulated both by ACC2 suppression and CPTI inhibitor treatment, which represent an altered fatty acid β-oxidation. And the knockdown of ACC2 reduced PA-induced autophagy and thus protects the cells from PA-induced lipotoxicity with attenuated lipid accumulation and rescued cell viability. Collectively, the present study proposed a novel autophagy-involved mechanism of PA-induced renal lipotoxicity and provided potential therapeutic strategy by modulating lipid β-oxidation for diabetic nephropathy.
Collapse
Affiliation(s)
- Wei Xin
- Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xu Zhao
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Lei Liu
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Ying Xu
- Department of Nephrology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, 250014, China
| | - Zhaoping Li
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Liyong Chen
- Department of Nutrition, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xiaojie Wang
- School of Medicine, Shandong University, Jinan, 250012, China
| | - Fan Yi
- Department of Pharmacology, Shandong University School of Medicine, Jinan, 250012, China
| | - Qiang Wan
- Department of Nephrology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, 250014, China.
| |
Collapse
|
145
|
Tong X, Ma H, Amadi SW, Ma L, Wu G. Reno-protection of G004, a novel anti-diabetic sulfonylurea in db/db mice. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:831-41. [PMID: 25943026 DOI: 10.1007/s00210-015-1112-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 02/24/2015] [Indexed: 12/26/2022]
Abstract
1-[4-[2-(4-Bromobenzene-sulfonamino)ethyl]phenylsulfonyl]-3-(trans-4-methylcyclohexyl) urea (G004, CAS865483-06-3) is a synthetic sulfonylurea, incorporating the hypoglycemic active structure of glimepiride (CAS 93479-97-1) and anti-TXA2 receptor (TP) active structure of BM-531(CAS 284464-46-6). In this study, we evaluated the effect of G004 on hyperglycemia and dyslipidemia as well as diabetic nephropathy (DN) in db/db mice by gavage over 90 consecutive days of treatment. The fasting blood glucose (FBG), glucose, and insulin tolerance as well as dyslipidemia were effectively ameliorated in db/db mice treated with G004. Interestingly, renal histological results of db/db mice revealed that G004 markedly reversed the expansion of mesangial extracellular matrix (ECM), the early hallmark of DN. Indeed, G004 treatment downregulated the renal expressions of type 4 collagen (Col IV) and transforming growth factor-β1 (TGF-β1) in db/db mice. In addition, imbalance in expressions of matrix metalloproteinase-9 (MMP-9) and its tissue inhibitor-1 (TIMP-1) in db/db mice kidneys was observed. However, G004 increased and decreased the expressions of MMP-9 and TIMP-1, respectively. It is well known that TGF-β pathway signaling plays an essential role in hyperglycemia-induced cell protein synthesis. On the other hand, MMP/TIMP system is responsible for the breakdown and turnover of ECM. Thus, we speculate that G004 possibly attenuated ECM accumulation via remodeling the synthesis and degradation of ECM component Col IV through modulation in TGF-β1 and MMP-9/TIMP-1 expressions in kidneys of db/db mice. Results from this study provide a strong rationale for G004 to be an efficient glucose-controlling agent with significant reno-protective properties.
Collapse
Affiliation(s)
- Xiaohui Tong
- China Pharmaceutical University, Nanjing, 210009, China
| | | | | | | | | |
Collapse
|
146
|
Broussard JL, Chapotot F, Abraham V, Day A, Delebecque F, Whitmore HR, Tasali E. Sleep restriction increases free fatty acids in healthy men. Diabetologia 2015; 58:791-8. [PMID: 25702040 PMCID: PMC4358810 DOI: 10.1007/s00125-015-3500-4] [Citation(s) in RCA: 101] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2014] [Accepted: 12/22/2014] [Indexed: 02/03/2023]
Abstract
AIMS/HYPOTHESIS Sleep loss is associated with insulin resistance and an increased risk for type 2 diabetes, yet underlying mechanisms are not understood. Elevation of circulating non-esterified (i.e. free) fatty acid (NEFA) concentrations can lead to insulin resistance and plays a central role in the development of metabolic diseases. Circulating NEFA in healthy individuals shows a marked diurnal variation with maximum levels occurring at night, yet the impact of sleep loss on NEFA levels across the 24 h cycle remains unknown. We hypothesised that sleep restriction would alter hormones that are known to stimulate lipolysis and lead to an increase in NEFA levels. METHODS We studied 19 healthy young men under controlled laboratory conditions with four consecutive nights of 8.5 h in bed (normal sleep) and 4.5 h in bed (sleep restriction) in randomised order. The 24 h blood profiles of NEFA, growth hormone (GH), noradrenaline (norepinephrine), cortisol, glucose and insulin were simultaneously assessed. Insulin sensitivity was estimated by a frequently sampled intravenous glucose tolerance test. RESULTS Sleep restriction relative to normal sleep resulted in increased NEFA levels during the nocturnal and early-morning hours. The elevation in NEFA was related to prolonged nocturnal GH secretion and higher early-morning noradrenaline levels. Insulin sensitivity was decreased after sleep restriction and the reduction in insulin sensitivity was correlated with the increase in nocturnal NEFA levels. CONCLUSIONS/INTERPRETATION Sleep restriction in healthy men results in increased nocturnal and early-morning NEFA levels, which may partly contribute to insulin resistance and the elevated diabetes risk associated with sleep loss.
Collapse
Affiliation(s)
- Josiane L Broussard
- Diabetes and Obesity Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, THA E104, Los Angeles, CA, 90048, USA,
| | | | | | | | | | | | | |
Collapse
|
147
|
Abu Bakar MH, Sarmidi MR, Cheng KK, Ali Khan A, Suan CL, Zaman Huri H, Yaakob H. Metabolomics – the complementary field in systems biology: a review on obesity and type 2 diabetes. MOLECULAR BIOSYSTEMS 2015; 11:1742-74. [DOI: 10.1039/c5mb00158g] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This paper highlights the metabolomic roles in systems biology towards the elucidation of metabolic mechanisms in obesity and type 2 diabetes.
Collapse
Affiliation(s)
- Mohamad Hafizi Abu Bakar
- Department of Bioprocess Engineering
- Faculty of Chemical Engineering
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Mohamad Roji Sarmidi
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
- Innovation Centre in Agritechnology for Advanced Bioprocessing (ICA)
| | - Kian-Kai Cheng
- Department of Bioprocess Engineering
- Faculty of Chemical Engineering
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Abid Ali Khan
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
- Department of Biosciences
| | - Chua Lee Suan
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| | - Hasniza Zaman Huri
- Department of Pharmacy
- Faculty of Medicine
- University of Malaya
- 50603 Kuala Lumpur
- Malaysia
| | - Harisun Yaakob
- Institute of Bioproduct Development
- Universiti Teknologi Malaysia
- 81310 Johor Bahru
- Malaysia
| |
Collapse
|
148
|
Constantinopoulos P, Michalaki M, Kottorou A, Habeos I, Psyrogiannis A, Kalfarentzos F, Kyriazopoulou V. Cortisol in tissue and systemic level as a contributing factor to the development of metabolic syndrome in severely obese patients. Eur J Endocrinol 2015; 172:69-78. [PMID: 25336506 DOI: 10.1530/eje-14-0626] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CONTEXT Adrenal and extra-adrenal cortisol production may be involved in the development of metabolic syndrome (MetS). OBJECTIVE To investigate the activity of the hypothalamic-pituitary-adrenal (HPA) axis and the expression of HSD11B1, nuclear receptor subfamily 3, group C, member 1 (glucocorticoid receptors) α (NR3C1α) and β (NR3C1β) in the liver, subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) of severely obese patients with and without MetS. METHODS The study included 37 severely obese patients (BMI ≥ 40 kg/m(2)), 19 with MetS (MetS+ group) and 18 without (MetS- group), studied before and during bariatric surgery. Before the day of surgery, urinary free cortisol (UFC) and diurnal variation of serum and salivary cortisol were estimated. During surgery, biopsies of the liver, VAT and SAT were obtained. The expression of HSD11B1, NR3C1α and NR3C1β was evaluated by RT-PCR. RESULTS UFC and area under the curve for 24-h profiles of serum and salivary cortisol were lower in the MetS- group. In the MetS- group, mRNA levels of HSD11B1 in liver exhibited a negative correlation with liver NR3C1α (LNR3C1α) and VAT expression of HSD11B1 was lower than the MetS+ group. CONCLUSIONS We observed a downregulation of the NR3C1α expression and lower VAT mRNA levels of HSD11B1 in the MetS- group, indicating a lower selective tissue cortisol production and action that could protect these patients from the metabolic consequences of obesity. In the MetS- group, a lower activity of the HPA axis was also detected. Taken together, cortisol in tissue and systematic level might play a role in the development of MetS in severely obese patients.
Collapse
Affiliation(s)
- Petros Constantinopoulos
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Marina Michalaki
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Anastasia Kottorou
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Ioannis Habeos
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Agathoklis Psyrogiannis
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Fotios Kalfarentzos
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| | - Venetsana Kyriazopoulou
- Division of EndocrinologyDiabetes and Metabolic Diseases, Department of Internal MedicineDivision of Nutritional Support and Morbid ObesityDepartment of SurgeryMolecular Oncology LaboratoryMedical School, University of Patras, 26500 Patras, Greece
| |
Collapse
|
149
|
Huynh J, Xiong G, Bentley-Lewis R. A systematic review of metabolite profiling in gestational diabetes mellitus. Diabetologia 2014; 57:2453-64. [PMID: 25193282 PMCID: PMC4221524 DOI: 10.1007/s00125-014-3371-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 08/14/2014] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Gestational diabetes mellitus is associated with adverse maternal and fetal outcomes during, as well as subsequent to, pregnancy, including increased risk of type 2 diabetes and cardiovascular disease. Because of the importance of early risk stratification in preventing these complications, improved first-trimester biomarker determination for diagnosing gestational diabetes would enhance our ability to optimise both maternal and fetal health. Metabolomic profiling, the systematic study of small molecule products of biochemical pathways, has shown promise in the identification of key metabolites associated with the pathogenesis of several metabolic diseases, including gestational diabetes. This article provides a systematic review of the current state of research on biomarkers and gestational diabetes and discusses the clinical relevance of metabolomics in the prediction, diagnosis and management of gestational diabetes. METHODS We conducted a systematic search of MEDLINE (PubMed) up to the end of February 2014 using the key term combinations of 'metabolomics,' 'metabonomics,' 'nuclear magnetic spectroscopy,' 'mass spectrometry,' 'metabolic profiling' and 'amino acid profile' combined (AND) with 'gestational diabetes'. Additional articles were identified through searching the reference lists from included studies. Quality assessment of included articles was conducted through the use of QUADOMICS. RESULTS This systematic review included 17 articles. The biomarkers most consistently associated with gestational diabetes were asymmetric dimethylarginine and NEFAs. After QUADOMICS analysis, 13 of the 17 included studies were classified as 'high quality'. CONCLUSIONS/INTERPRETATION Existing metabolomic studies of gestational diabetes present inconsistent findings regarding metabolite profile characteristics. Further studies are needed in larger, more racially/ethnically diverse populations.
Collapse
Affiliation(s)
- Jennifer Huynh
- Department of Medicine/Diabetes Unit, Massachusetts General Hospital, 55 Fruit Street, Bulfinch 4-415, Boston, MA, 02114, USA
| | | | | |
Collapse
|
150
|
Malin SK, Kashyap SR, Hammel J, Miyazaki Y, DeFronzo RA, Kirwan JP. Adjusting glucose-stimulated insulin secretion for adipose insulin resistance: an index of β-cell function in obese adults. Diabetes Care 2014; 37:2940-6. [PMID: 25139885 PMCID: PMC4207203 DOI: 10.2337/dc13-3011] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE The hyperbolic relationship between insulin secretion and sensitivity has been used to assess in vivo β-cell function (i.e., the disposition index). The disposition index emphasizes the importance of taking into account both skeletal muscle and hepatic insulin resistance to depict insulin secretion. However, we propose that adipose tissue insulin resistance also needs to be accounted for when characterizing glucose-stimulated insulin secretion (GSIS) because elevated plasma free fatty acids (FFAs) impair β-cell function. RESEARCH DESIGN AND METHODS To characterize the adipose disposition index, we used [1-(14)C] palmitate infusion to determine basal FFA turnover rate/adipose insulin resistance and an oral glucose tolerance test to characterize the first (i.e., 0-30 min) and second phase (i.e., 60-120 min) of GSIS. We validated a simplified version of the tracer infusion calculation as the product of (1/plasma FFA concentration × plasma insulin concentration) × GSIS in 44 obese insulin-resistant subjects. RESULTS The plasma FFA and palmitate tracer infusion calculations of the first- and second-phase disposition index were strongly correlated (r = 0.86, P < 0.000001 and r = 0.89, P < 0.000001, respectively). The first- and second-phase adipose disposition index derived from plasma FFA also was tightly associated with fasting hyperglycemia (r = -0.87, P < 0.00001 and r = -0.89, P < 0.00001, respectively) and 2-h glucose concentrations (r = -0.86, P < 0.00001 and r = -0.90, P < 0.00001). CONCLUSIONS Adjusting GSIS for adipose insulin resistance provides an index of β-cell function in obese subjects across the glucose spectrum. Plasma FFA-derived calculations of β-cell function may provide additional insight into the role of adipose tissue in glucose regulation.
Collapse
Affiliation(s)
- Steven K Malin
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH
| | - Sangeeta R Kashyap
- Department of Endocrinology, Diabetes, and Metabolism, Cleveland Clinic, Cleveland, OH
| | - Jeff Hammel
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Yoshi Miyazaki
- University of Texas Health Science Center, Diabetes Division, San Antonio, TX
| | - Ralph A DeFronzo
- University of Texas Health Science Center, Diabetes Division, San Antonio, TX
| | - John P Kirwan
- Department of Pathobiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, OH Metabolic Translational Research Center, Endocrine and Metabolism Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|