101
|
Badimon L, Bugiardini R, Cenko E, Cubedo J, Dorobantu M, Duncker DJ, Estruch R, Milicic D, Tousoulis D, Vasiljevic Z, Vilahur G, de Wit C, Koller A. Position paper of the European Society of Cardiology-working group of coronary pathophysiology and microcirculation: obesity and heart disease. Eur Heart J 2019; 38:1951-1958. [PMID: 28873951 DOI: 10.1093/eurheartj/ehx181] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 04/28/2017] [Indexed: 12/15/2022] Open
Affiliation(s)
- Lina Badimon
- Cardiovascular Research Center (CSIC-ICCC), CIBERCV, and Biomedical Research Institute Sant Pau (IIB-Sant Pau), c/Sant Antoni M Claret 167, 08025 Barcelona, Spain.,Cardiovascular Research Chair UAB, Barcelona, Spain
| | - Raffaele Bugiardini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Giuseppe Massarenti 9, 40138 Bologna, Italy
| | - Edina Cenko
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Via Giuseppe Massarenti 9, 40138 Bologna, Italy
| | - Judit Cubedo
- Cardiovascular Research Center (CSIC-ICCC), CIBERCV, and Biomedical Research Institute Sant Pau (IIB-Sant Pau), c/Sant Antoni MaClaret 167, 08025 Barcelona, Spain
| | - Maria Dorobantu
- Cardiology Department, University of Medicine and Pharmacy "Carol Davila" of Bucharest, Emergency Clinical Hospital of Bucharest, 8, Calea Floreasca, Sector 1, 014461 Bucuresti, Romania
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Cardiovascular Research Institute COEUR, Erasmus University Medical Center, PO Box 1738, 3000 DR, Rotterdam, The Netherlands
| | - Ramón Estruch
- Department of Internal Medicine, Hospital Clinic, IDIBAPS, University of Barcelona, Villarroel, 170, 08036 Barcelona, Spain.,CIBER Obesity and Nutrition, Instituto de Salud Carlos III, Spain
| | - Davor Milicic
- Department for Cardiovascular Diseases, University Hospital Center Zagreb, University of Zagreb, Kispaticeva 12, HR-10000 Zagreb, Croatia
| | - Dimitris Tousoulis
- First Department of Cardiology, Hippokration Hospital, University of Athens Medical School, Vasilissis Sofias 114, TK 115 28 Athens, Greece
| | - Zorana Vasiljevic
- Clinical Center of Serbia, University of Belgrade, Pasterova 2, 11000 Belgrade, Serbia
| | - Gemma Vilahur
- Cardiovascular Research Center (CSIC-ICCC), CIBERCV, and Biomedical Research Institute Sant Pau (IIB-Sant Pau), c/Sant Antoni MaClaret 167, 08025 Barcelona, Spain
| | - Cor de Wit
- Institut für Physiologie, Universität zu Lübeck and Deutsches Zentrumfür Herz-Kreislauf-Forschung (DZHK) e.V., partner site: Hamburg/Kiel/Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Akos Koller
- Institute of Natural Sciences, University of Physical Education, Alkotas street, 44, 1123 Budapest, Hungary.,Department of Physiology, New York Medical College, Valhalla, NY 10595, USA
| |
Collapse
|
102
|
Singla R, Murthy M, Singla S, Gupta Y. Friendly Fat Theory - Explaining the Paradox of Diabetes and Obesity. EUROPEAN ENDOCRINOLOGY 2019; 15:25-28. [PMID: 31244907 PMCID: PMC6587901 DOI: 10.17925/ee.2019.15.1.25] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022]
Abstract
Obesity has been called the mother of all diseases and, historically, has been strongly linked to diabetes. However, there are still some paradoxes that exist in diabetes epidemiology and obesity and no unifying hypothesis has been proposed to explain these paradoxical phenomena. Despite the ever-increasing prevalence of both obesity and diabetes, differential relationships exist between diabetes and the extent of obesity in various different ethnic groups. In addition, people with a higher body mass index have been shown to have an improved survival advantage in terms of chronic diabetes complications, especially cardiovascular complications. This narrative review attempts to explain these paradoxical and complex relationships with a single unifying theory. We propose that adipocytes are actually friends of the human body to prevent the occurrence of diabetes and also help in mitigating the complications of diabetes. Adipose tissue actually acts as a reservoir of free fatty acids, responsible for insulin resistance, and prevents their overflow into insulin-sensitive tissues and, therefore, friendly fat theory.
Collapse
Affiliation(s)
- Rajiv Singla
- Department of Endocrinology, Kalpavriksh Superspeciality Centre, Delhi
| | - Mithun Murthy
- Warrington and Halton NHS Foundation Trust Warrington Hospital, Warrington
| | - Sweta Singla
- Department of Neurology, Kalpavriksh Superspeciality Centre, Delhi
| | - Yashdeep Gupta
- Department of Endocrinology, All India Institute of Medical Sciences, Delhi
| |
Collapse
|
103
|
Dyer DP, Nebot JB, Kelly CJ, Medina‐Ruiz L, Schuette F, Graham GJ. The chemokine receptor CXCR2 contributes to murine adipocyte development. J Leukoc Biol 2019; 105:497-506. [PMID: 30517976 PMCID: PMC6392114 DOI: 10.1002/jlb.1a0618-216rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 11/16/2018] [Accepted: 11/18/2018] [Indexed: 01/20/2023] Open
Abstract
Chemokines are members of a large family of chemotactic cytokines that signal through their receptors to mediate leukocyte recruitment during inflammation and homeostasis. The chemokine receptor CXCR2 has largely been associated with neutrophil recruitment. However, there is emerging evidence of roles for chemokines and their receptors in processes other than leukocyte migration. We have previously demonstrated that CXCR2 knockout (KO) mice have thinner skin compared to wild-type mice. Herein we demonstrate that this is due to a thinner subcutaneous adipose layer, as a result of fewer and smaller individual adipocytes. We observe a similar phenotype in other fat depots and present data that suggests this may be due to reduced expression of adipogenesis related genes associated with adipocyte specific CXCR2 signaling. Interestingly, this phenotype is evident in female, but not male, CXCR2 KO mice. These findings expand our understanding of nonleukocyte related chemokine receptor functions and help to explain some previously observed adipose-related phenotypes in CXCR2 KO mice.
Collapse
Affiliation(s)
- Douglas P. Dyer
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
- Wellcome Centre for Cell‐Matrix Research, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUnited Kingdom
- Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, Manchester Academic Health Science CentreUniversity of ManchesterManchesterUnited Kingdom
| | - Joan Boix Nebot
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
| | - Christopher J. Kelly
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
| | - Laura Medina‐Ruiz
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
| | - Fabian Schuette
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
| | - Gerard J Graham
- Chemokine Research Group, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life SciencesUniversity of GlasgowGlasgowUnited Kingdom
| |
Collapse
|
104
|
Transcriptional Regulation of Acyl-CoA:Glycerol- sn-3-Phosphate Acyltransferases. Int J Mol Sci 2019; 20:ijms20040964. [PMID: 30813330 PMCID: PMC6412627 DOI: 10.3390/ijms20040964] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 02/14/2019] [Accepted: 02/15/2019] [Indexed: 12/13/2022] Open
Abstract
Acyl-CoA:glycerol-sn-3-phosphate acyltransferase (GPAT) is an enzyme responsible for the rate-limiting step in the synthesis of glycerophospholipids and triacylglycerol (TAG). The enzymes of mammalian species are classified into four isoforms; GPAT1 and GPAT2 are localized in the mitochondrial outer membrane, whereas GPAT3 and GPAT4 are localized in the endoplasmic reticulum membrane. The activity of each enzyme expressed is associated with physiological and pathological functions. The transcriptional regulation is well known, particularly in GPAT1. GPAT1 mRNA expression is mainly regulated by the binding of the transcriptional factor SREBP-1c to the specific element (the sterol regulatory element) flanking the GPAT1 promoter. The TAG level is controlled by the insulin-induced transcriptional expression of GPAT1, which occupies most of the GPAT activity in the liver. The transcriptional regulation of the other three GPAT isoforms remains undetermined in detail. It is predicted that retinoic acid serves as a transcription factor in the GPAT2 promoter. PPARγ (peroxisome proliferator-activated receptor γ) increases the mRNA expression of GPAT3, which is associated with TAG synthesis in adipose tissues. Although GPAT has been considered to be a key enzyme in the production of TAG, unexpected functions have recently been reported, particularly in GPAT2. It is likely that GPAT2 is associated with tumorigenesis and normal spermatogenesis. In this review, the physiological and pathophysiological roles of the four GPAT isoforms are described, alongside the transcriptional regulation of these enzymes.
Collapse
|
105
|
Insulin and Insulin Receptors in Adipose Tissue Development. Int J Mol Sci 2019; 20:ijms20030759. [PMID: 30754657 PMCID: PMC6387287 DOI: 10.3390/ijms20030759] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Insulin is a major endocrine hormone also involved in the regulation of energy and lipid metabolism via the activation of an intracellular signaling cascade involving the insulin receptor (INSR), insulin receptor substrate (IRS) proteins, phosphoinositol 3-kinase (PI3K) and protein kinase B (AKT). Specifically, insulin regulates several aspects of the development and function of adipose tissue and stimulates the differentiation program of adipose cells. Insulin can activate its responses in adipose tissue through two INSR splicing variants: INSR-A, which is predominantly expressed in mesenchymal and less-differentiated cells and mainly linked to cell proliferation, and INSR-B, which is more expressed in terminally differentiated cells and coupled to metabolic effects. Recent findings have revealed that different distributions of INSR and an altered INSR-A:INSR-B ratio may contribute to metabolic abnormalities during the onset of insulin resistance and the progression to type 2 diabetes. In this review, we discuss the role of insulin and the INSR in the development and endocrine activity of adipose tissue and the pharmacological implications for the management of obesity and type 2 diabetes.
Collapse
|
106
|
Iyengar NM, Arthur R, Manson JE, Chlebowski RT, Kroenke CH, Peterson L, Cheng TYD, Feliciano EC, Lane D, Luo J, Nassir R, Pan K, Wassertheil-Smoller S, Kamensky V, Rohan TE, Dannenberg AJ. Association of Body Fat and Risk of Breast Cancer in Postmenopausal Women With Normal Body Mass Index: A Secondary Analysis of a Randomized Clinical Trial and Observational Study. JAMA Oncol 2019; 5:155-163. [PMID: 30520976 PMCID: PMC6439554 DOI: 10.1001/jamaoncol.2018.5327] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/30/2018] [Indexed: 01/06/2023]
Abstract
Importance Obesity is associated with an increased risk of breast cancer, including the estrogen receptor (ER)-positive subtype in postmenopausal women. Whether excess adiposity is associated with increased risk in women with a normal body mass index (BMI; calculated as weight in kilograms divided by height in meters squared) is unknown. Objective To investigate the association between body fat and breast cancer risk in women with normal BMI. Design, Setting, and Participants This ad hoc secondary analysis of the Women's Health Initiative (WHI) clinical trial and observational study cohorts was restricted to postmenopausal participants with a BMI ranging from 18.5 to 24.9. Women aged 50 to 79 years were enrolled from October 1, 1993, through December 31, 1998. Of these, 3460 participants underwent body fat measurement with dual-energy x-ray absorptiometry (DXA) at 3 US designated centers with follow-up. At a median follow-up of 16 years (range, 9-20 years), 182 incident breast cancers had been ascertained, and 146 were ER positive. Follow-up was complete on September 30, 2016, and data from October 1, 1993, through September 30, 2016, was analyzed August 2, 2017, through August 21, 2018. Main Outcomes and Measures Body fat levels were measured at baseline and years 1, 3, 6, and 9 using DXA. Information on demographic data, medical history, and lifestyle factors was collected at baseline. Invasive breast cancers were confirmed via central review of medical records by physician adjudicators. Blood analyte levels were measured in subsets of participants. Results Among the 3460 women included in the analysis (mean [SD] age, 63.6 [7.6] years), multivariable-adjusted hazard ratios for the risk of invasive breast cancer were 1.89 (95% CI, 1.21-2.95) for the highest quartile of whole-body fat and 1.88 (95% CI, 1.18-2.98) for the highest quartile of trunk fat mass. The corresponding adjusted hazard ratios for ER-positive breast cancer were 2.21 (95% CI, 1.23-3.67) and 1.98 (95% CI, 1.18-3.31), respectively. Similar positive associations were observed for serial DXA measurements in time-dependent covariate analyses. Circulating levels of insulin, C-reactive protein, interleukin 6, leptin, and triglycerides were higher, whereas levels of high-density lipoprotein cholesterol and sex hormone-binding globulin were lower in those in the uppermost vs lowest quartiles of trunk fat mass. Conclusions and Relevance In postmenopausal women with normal BMI, relatively high body fat levels were associated with an elevated risk of invasive breast cancer and altered levels of circulating metabolic and inflammatory factors. Normal BMI categorization may be an inadequate proxy for the risk of breast cancer in postmenopausal women. Trial Registration ClinicalTrials.gov identifier: NCT00000611.
Collapse
Affiliation(s)
- Neil M. Iyengar
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
- Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Rhonda Arthur
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - JoAnn E. Manson
- Division of Preventive Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Rowan T. Chlebowski
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, California
| | | | - Lindsay Peterson
- Department of Medicine, Washington University in Saint Louis, St Louis, Missouri
| | | | | | - Dorothy Lane
- Department of Family, Population and Preventive Medicine, Stony Brook University School of Medicine, Stony Brook, New York
| | - Juhua Luo
- Department of Epidemiology and Biostatistics, Indiana University, Indianapolis
| | - Rami Nassir
- Department of Biochemistry and Molecular Medicine, University of California, Davis
| | - Kathy Pan
- Los Angeles Biomedical Research Institute at Harbor-UCLA (University of California, Los Angeles) Medical Center, Los Angeles
| | | | - Victor Kamensky
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Thomas E. Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | | |
Collapse
|
107
|
Zhao L, Zhu C, Lu M, Chen C, Nie X, Abudukerimu B, Zhang K, Ning Z, Chen Y, Cheng J, Xia F, Wang N, Jensen MD, Lu Y. The key role of a glucagon-like peptide-1 receptor agonist in body fat redistribution. J Endocrinol 2019; 240:271-286. [PMID: 30530905 DOI: 10.1530/joe-18-0374] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 11/08/2018] [Indexed: 01/01/2023]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1RAs) are an ideal therapy for type 2 diabetes and, as of recently, for obesity. In contrast to visceral fat, subcutaneous fat appears to be protective against metabolic diseases. Here, we aimed to explore whether liraglutide, a GLP-1RA, could redistribute body fat via regulating lipid metabolism in different fat depots. After being fed a high-fat diet for 8 weeks, 50 male Wistar and Goto-Kakizaki rats were randomly divided into a normal control group, a diabetic control group, low- and high-dose liraglutide-treated groups and a diet-control group. Different doses of liraglutide (400 μg/kg/day or 1200 μg/kg/day) or an equal volume of normal saline were administered to the rats subcutaneously once a day for 12 weeks. Body composition and body fat deposition were measured by dual-energy X-ray absorptiometry and MRI. Isotope tracers were infused to explore lipid metabolism in different fat depots. Quantitative real-time PCR and Western blot analyses were conducted to evaluate the expression of adipose-related genes. The results showed that liraglutide decreased visceral fat and relatively increased subcutaneous fat. Lipogenesis was reduced in visceral white adipose tissue (WAT) but was elevated in subcutaneous WAT. Lipolysis was also attenuated, and fatty acid oxidation was enhanced. The mRNA expression levels of adipose-related genes in different tissues displayed similar trends after liraglutide treatment. In addition, the expression of browning-related genes was upregulated in subcutaneous WAT. Taken together, the results suggested that liraglutide potentially redistributes body fat and promotes browning remodeling in subcutaneous WAT to improve metabolic disorders.
Collapse
Affiliation(s)
- Li Zhao
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chunfang Zhu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Meng Lu
- Research Center for Clinical Medicine, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaomin Nie
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Buatikamu Abudukerimu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Kun Zhang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Zhiyuan Ning
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Jing Cheng
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | | | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| |
Collapse
|
108
|
Macdougall CE, Longhi MP. Adipose tissue dendritic cells in steady-state. Immunology 2019; 156:228-234. [PMID: 30552824 DOI: 10.1111/imm.13034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 11/21/2018] [Accepted: 11/28/2018] [Indexed: 12/17/2022] Open
Abstract
Healthy white adipose tissue (WAT) participates in regulating systemic metabolism, whereas dysfunctional WAT plays a prominent role in the development of obesity-associated co-morbidities. Tissue-resident immune cells are important for maintaining WAT homeostasis, including conventional dendritic cells (cDCs) which are critical in the initiation and regulation of adaptive immune responses. Due to phenotypic overlap with other myeloid cells, the distinct contribution of WAT cDCs has been poorly understood. This review will discuss the contribution of cDCs in the maintenance of WAT homeostasis. In particular, the review will focus on the metabolic cross-talk between cDCs and adipocytes that regulates local immune responses during physiological conditions.
Collapse
Affiliation(s)
- Claire E Macdougall
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| | - M Paula Longhi
- William Harvey Research Institute, Barts and the London, Queen Mary University of London, London, UK
| |
Collapse
|
109
|
Andrade EF, de Oliveira Silva V, Orlando DR, Pereira LJ. Mechanisms Involved in Glycemic Control Promoted by Exercise in Diabetics. Curr Diabetes Rev 2019; 15:105-110. [PMID: 29446746 DOI: 10.2174/1573399814666180214144717] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/22/2018] [Accepted: 01/31/2018] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Diabetes mellitus is a metabolic disease characterized by high glycemic levels for long periods. This disease has a high prevalence in the world population, being currently observed an increase in its incidence. This fact is mainly due to the sedentary lifestyle and hypercaloric diets. Non-pharmacological interventions for glycemic control include exercise, which promotes changes in skeletal muscle and adipocytes. Thus, increased glucose uptake by skeletal muscle and decreased insulin resistance through modulating adipocytes are the main factors that improve glycemic control against diabetes. CONCLUSION It was sought to elucidate mechanisms involved in the improvement of glycemic control in diabetics in front of the exercise.
Collapse
Affiliation(s)
| | | | - Débora Ribeiro Orlando
- Department of Agricultural Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Unai, Brazil
| | | |
Collapse
|
110
|
Gancheva S, Jelenik T, Álvarez-Hernández E, Roden M. Interorgan Metabolic Crosstalk in Human Insulin Resistance. Physiol Rev 2018; 98:1371-1415. [PMID: 29767564 DOI: 10.1152/physrev.00015.2017] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Excessive energy intake and reduced energy expenditure drive the development of insulin resistance and metabolic diseases such as obesity and type 2 diabetes mellitus. Metabolic signals derived from dietary intake or secreted from adipose tissue, gut, and liver contribute to energy homeostasis. Recent metabolomic studies identified novel metabolites and enlarged our knowledge on classic metabolites. This review summarizes the evidence of their roles as mediators of interorgan crosstalk and regulators of insulin sensitivity and energy metabolism. Circulating lipids such as free fatty acids, acetate, and palmitoleate from adipose tissue and short-chain fatty acids from the gut effectively act on liver and skeletal muscle. Intracellular lipids such as diacylglycerols and sphingolipids can serve as lipotoxins by directly inhibiting insulin action in muscle and liver. In contrast, fatty acid esters of hydroxy fatty acids have been recently shown to exert a series of beneficial effects. Also, ketoacids are gaining interest as potent modulators of insulin action and mitochondrial function. Finally, branched-chain amino acids not only predict metabolic diseases, but also inhibit insulin signaling. Here, we focus on the metabolic crosstalk in humans, which regulates insulin sensitivity and energy homeostasis in the main insulin-sensitive tissues, skeletal muscle, liver, and adipose tissue.
Collapse
Affiliation(s)
- Sofiya Gancheva
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Tomas Jelenik
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Elisa Álvarez-Hernández
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| | - Michael Roden
- Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University , Düsseldorf , Germany ; Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University , Düsseldorf , Germany ; and German Center of Diabetes Research (DZD e.V.), Munich- Neuherberg , Germany
| |
Collapse
|
111
|
Kaisanlahti A, Glumoff T. Browning of white fat: agents and implications for beige adipose tissue to type 2 diabetes. J Physiol Biochem 2018; 75:1-10. [PMID: 30506389 PMCID: PMC6513802 DOI: 10.1007/s13105-018-0658-5] [Citation(s) in RCA: 152] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/31/2018] [Indexed: 12/23/2022]
Abstract
Mammalian adipose tissue is traditionally categorized into white and brown relating to their function and morphology: while white serves as an energy storage, brown adipose tissue acts as the heat generator maintaining the core body temperature. The most recently identified type of fat, beige adipocyte tissue, resembles brown fat by morphology and function but is developmentally more related to white. The synthesis of beige fat, so-called browning of white fat, has developed into a topical issue in diabetes and metabolism research. This is due to its favorable effect on whole-body energy metabolism and the fact that it can be recruited during adult life. Indeed, brown and beige adipose tissues have been demonstrated to play a role in glucose homeostasis, insulin sensitivity, and lipid metabolism—all factors related to pathogenesis of type 2 diabetes. Many agents capable of initiating browning have been identified so far and tested widely in humans and animal models including in vitro and in vivo experiments. Interestingly, several agents demonstrated to have browning activity are in fact secreted as adipokines from brown and beige fat tissue, suggesting a physiological relevance both in beige adipocyte recruitment processes and in maintenance of metabolic homeostasis. The newest findings on agents driving beige fat recruitment, their mechanisms, and implications on type 2 diabetes are discussed in this review.
Collapse
MESH Headings
- Adipose Tissue, Beige/drug effects
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, Beige/pathology
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, Brown/pathology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/pathology
- Animals
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/metabolism
- Diabetes Mellitus, Type 2/pathology
- Energy Metabolism/drug effects
- Energy Metabolism/genetics
- Glucagon-Like Peptide 1/pharmacology
- Glucose/metabolism
- Humans
- Insulin Resistance
- Leptin/pharmacology
- Lipid Metabolism/drug effects
- Lipid Metabolism/genetics
- Lipotropic Agents/pharmacology
- Melatonin/pharmacology
- Natriuretic Peptides/pharmacology
- Thermogenesis/drug effects
- Thermogenesis/genetics
- Tretinoin/pharmacology
Collapse
Affiliation(s)
- A Kaisanlahti
- Biocenter Oulu/Cancer Research and Translational Medicine Research Unit, University of Oulu, Aapistie 5, P.O. Box 5281, 90014, Oulu, Finland.
| | - T Glumoff
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7A, P.O Box 5400, 90014, Oulu, Finland
| |
Collapse
|
112
|
Druzhilov MA, Kuznetsova ТY, Druzhilova ОY. “Obesity paradoxes”: main causes of an “inverse” cardiovascular epidemiology. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2018. [DOI: 10.15829/1728-8800-2018-5-92-98] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Taken the continuous increase of obesity prevalence, most countries in the world deal with an epidemy, one of the main healthcare concerns. In the structure of nosology associated with overweight and obesity, cardiovascular is leading. Also, in the recent trials and meta analyses there is negative correlation found for body mass index and clinical outcomes characterizing better survival and lower events rate in those with higher BMI, as less chronic diseases. Such facts facilitated a number of discussions on the predictive value of overweight and obesity and consideration whether to correct those in cardiovascular patients as secondary prevention. Current article is focused on the main causes for an “inverse” cardiovascular epidemiology in overweight and obesity.
Collapse
Affiliation(s)
- M. A. Druzhilov
- Medical and Sanitary Institution of the FSS in Karelia Republic
| | | | | |
Collapse
|
113
|
Katsiki N, Mikhailidis DP, Mantzoros C. Non-alcoholic fatty liver disease and colorectal cancer: A marker of risk or common causation? Metabolism 2018; 87:A10-A13. [PMID: 30172755 DOI: 10.1016/j.metabol.2018.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Niki Katsiki
- Second Propedeutic Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippocration Hospital, Thessaloniki, Greece.
| | - Dimitri P Mikhailidis
- Department of Clinical Biochemistry, Royal Free Hospital campus, University College London Medical School, University College London (UCL), London, UK
| | - Christos Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
114
|
Pi-Sunyer X. Changes in body composition and metabolic disease risk. Eur J Clin Nutr 2018; 73:231-235. [PMID: 30275524 DOI: 10.1038/s41430-018-0320-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/06/2018] [Indexed: 11/09/2022]
Abstract
As individuals gain weight, they increase the amount of fat that they accrue on their body. This causes adipocytes to enlarge and increases not only subcutaneous fat but also deposits fat in other vulnerable areas of the body. This ectopic fat is deposited in the intra-abdominal visceral fat depot, in muscle, in the liver and in the beta cells. Fat in these locations initiates a dysfunctional state in these insulin-sensitive tissues leading to insulin resistance, the appearance of the Metabolic Syndrome, and an increased risk of developing both type 2 diabetes and cardiovascular disease. A loss of weight and with it a loss of fat decreases this risk.
Collapse
Affiliation(s)
- Xavier Pi-Sunyer
- Columbia University College of Physicians and Surgeons and Columbia Institute of Human Nutrition, New York, NY, USA.
| |
Collapse
|
115
|
Vassilatou E, Lafoyianni S, Vassiliadi DA, Ioannidis D, Paschou SA, Mizamtsidi M, Panagou M, Vryonidou A. Visceral adiposity index for the diagnosis of nonalcoholic fatty liver disease in premenopausal women with and without polycystic ovary syndrome. Maturitas 2018; 116:1-7. [DOI: 10.1016/j.maturitas.2018.06.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/27/2018] [Accepted: 06/30/2018] [Indexed: 12/18/2022]
|
116
|
Trouwborst I, Bowser SM, Goossens GH, Blaak EE. Ectopic Fat Accumulation in Distinct Insulin Resistant Phenotypes; Targets for Personalized Nutritional Interventions. Front Nutr 2018; 5:77. [PMID: 30234122 PMCID: PMC6131567 DOI: 10.3389/fnut.2018.00077] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/15/2018] [Indexed: 12/13/2022] Open
Abstract
Cardiometabolic diseases are one of the leading causes for disability and mortality in the Western world. The prevalence of these chronic diseases is expected to rise even further in the next decades. Insulin resistance (IR) and related metabolic disturbances are linked to ectopic fat deposition, which is the storage of excess lipids in metabolic organs such as liver and muscle. Notably, a vicious circle exists between IR and ectopic fat, together increasing the risk for the development of cardiometabolic diseases. Nutrition is a key-determining factor for both IR and ectopic fat deposition. The macronutrient composition of the diet may impact metabolic processes related to ectopic fat accumulation and IR. Interestingly, however, the metabolic phenotype of an individual may determine the response to a certain diet. Therefore, population-based nutritional interventions may not always lead to the most optimal (cardiometabolic) outcomes at the individual level, and differences in the metabolic phenotype may underlie conflicting findings related to IR and ectopic fat in dietary intervention studies. Detailed metabolic phenotyping will help to better understand the complex relationship between diet and metabolic regulation, and to optimize intervention outcomes. A subgroup-based approach that integrates, among others, tissue-specific IR, cardiometabolic parameters, anthropometrics, gut microbiota, age, sex, ethnicity, and psychological factors may thereby increase the efficacy of dietary interventions. Nevertheless, the implementation of more personalized nutrition may be complex, costly, and time consuming. Future studies are urgently warranted to obtain insight into a more personalized approach to nutritional interventions, taking into account the metabolic phenotype to ultimately improve insulin sensitivity and reduce the risk for cardiometabolic diseases.
Collapse
Affiliation(s)
- Inez Trouwborst
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Suzanne M Bowser
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Ellen E Blaak
- Department of Human Biology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
117
|
A selective inhibitor of ceramide synthase 1 reveals a novel role in fat metabolism. Nat Commun 2018; 9:3165. [PMID: 30131496 PMCID: PMC6104039 DOI: 10.1038/s41467-018-05613-7] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 07/17/2018] [Indexed: 02/06/2023] Open
Abstract
Specific forms of the lipid ceramide, synthesized by the ceramide synthase enzyme family, are believed to regulate metabolic physiology. Genetic mouse models have established C16 ceramide as a driver of insulin resistance in liver and adipose tissue. C18 ceramide, synthesized by ceramide synthase 1 (CerS1), is abundant in skeletal muscle and suggested to promote insulin resistance in humans. We herein describe the first isoform-specific ceramide synthase inhibitor, P053, which inhibits CerS1 with nanomolar potency. Lipidomic profiling shows that P053 is highly selective for CerS1. Daily P053 administration to mice fed a high-fat diet (HFD) increases fatty acid oxidation in skeletal muscle and impedes increases in muscle triglycerides and adiposity, but does not protect against HFD-induced insulin resistance. Our inhibitor therefore allowed us to define a role for CerS1 as an endogenous inhibitor of mitochondrial fatty acid oxidation in muscle and regulator of whole-body adiposity. Ceramides are signalling molecules that regulate several physiological functions including insulin sensitivity. Here the authors report a selective ceramide synthase 1 inhibitor that counteracts lipid accumulation within the muscle and adiposity by increasing fatty acid oxidation but without affecting insulin sensitivity in mice fed with an obesogenic diet.
Collapse
|
118
|
Lee DH, Keum N, Hu FB, Orav EJ, Rimm EB, Willett WC, Giovannucci EL. Comparison of the association of predicted fat mass, body mass index, and other obesity indicators with type 2 diabetes risk: two large prospective studies in US men and women. Eur J Epidemiol 2018; 33:1113-1123. [PMID: 30117031 DOI: 10.1007/s10654-018-0433-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 08/10/2018] [Indexed: 02/06/2023]
Abstract
Obesity, defined by body mass index (BMI), is a well-established risk factor of type 2 diabetes, but BMI has been criticized for its inability to discriminate fat mass and lean body mass. We examined the association between predicted fat mass and type 2 diabetes risk in two large US prospective cohorts, and compared the magnitude of the association with BMI and other obesity indicators. Validated anthropometric prediction equations previously developed from the National Health and Nutrition Examination Survey were used to estimate predicted fat mass and percent fat for 97,111 participants from the Health Professionals Follow-up Study (1987-2012) and the Nurses' Health Study (1986-2012) who were followed up for type 2 diabetes. Multivariable-adjusted hazard ratios for type 2 diabetes across quintiles of predicted fat mass were 1.00, 1.96, 2.96, 3.90, and 8.38 for men and 1.00, 2.20, 3.50, 5.73, and 12.1 for women; of BMI were 1.00, 1.69, 2.45, 3.54, and 6.94 for men and 1.00, 1.76, 2.86, 4.88, and 9.88 for women. Predicted FM showed the strongest association with type 2 diabetes in men followed by waist circumference (WC), waist-to-height ratio (WHtR), predicted percent fat, BMI, Waist-to-hip ratio (WHR), and a body shape index (ABSI). For women, the strongest association was shown for WHtR, followed by WC, predicted percent fat, predicted fat mass, BMI, ABSI, and WHR. Compared to BMI, predicted fat mass demonstrated consistently stronger association with type 2 diabetes risk. However, there was inconclusive evidence to suggest that predicted fat mass is substantially superior to other obesity indicators.
Collapse
Affiliation(s)
- Dong Hoon Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA
| | - NaNa Keum
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.,Department of Food Science and Biotechnology, Dongguk University, Goyang, South Korea
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - E John Orav
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.,Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Edward L Giovannucci
- Department of Nutrition, Harvard T.H. Chan School of Public Health, 665 Huntington Avenue, Boston, MA, 02115, USA. .,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA. .,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
119
|
Trinh L, Lind E, Peterson P, Svensson J, Olsson LE, Månsson S. High-Resolution MR Imaging of Muscular Fat Fraction-Comparison of Three T 2-Based Methods and Chemical Shift-Encoded Imaging. Tomography 2018; 3:153-162. [PMID: 30042979 PMCID: PMC6024436 DOI: 10.18383/j.tom.2017.00011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Chemical shift-encoded imaging (CSEI) is the most common magnetic resonance imaging fat–water separation method. However, when high spatial resolution fat fraction (FF) images are desired, CSEI might be challenging owing to the increased interecho spacing. Here, 3 T2-based methods have been assessed as alternative methods for obtaining high-resolution FF images. Images from the calf of 10 healthy volunteers were acquired; FF maps were then estimated using 3 T2-based methods (2- and 3-parameter nonlinear least squares fit and a Bayesian probability method) and CSEI for reference. In addition, simulations were conducted to characterize the performance of various methods. Here, all T2-based methods resulted in qualitatively improved high-resolution FF images compared with high-resolution CSEI. The 2-parameter fit showed best quantitative agreement to low-resolution CSEI, even at low FF. The estimated T2-values of fat and water, and the estimated muscle FF of the calf, agreed well with previously published data. In conclusion, T2-based methods can provide improved high-resolution FF images of the calf compared with the CSEI method.
Collapse
Affiliation(s)
- Lena Trinh
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Emelie Lind
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden.,Department of Medical Radiation Physics, Lund University, Lund, Sweden
| | - Pernilla Peterson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Jonas Svensson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden.,Medical Imaging and Physiology, Skåne University Hospital, Lund, Sweden
| | - Lars E Olsson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Sven Månsson
- Medical Radiation Physics, Department of Translational Medicine, Lund University, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
120
|
Inactivation of MAPK in epididymal fat and amelioration of triglyceride secretion by injection of GRK2 siRNA in ob/ob mice. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:1075-1083. [PMID: 29946903 DOI: 10.1007/s00210-018-1530-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023]
Abstract
Abnormal G protein-coupled receptor kinase 2 (GRK2) accumulation has a crucial role in the development of insulin resistance and diabetes. Although GRK2 siRNA transfection in the liver improves insulin resistance-related vascular complications, the effects of GRK2 siRNA in lipid metabolism and obesity remain unknown. To investigate how GRK2 siRNA affects obesity, ob/ob mice were transfected with GRK2 siRNA, mainly in the liver, by using a hydrodynamic-based procedure. Epididymal fat, glucose, triglyceride, non-esterified fatty acid (NEFA), and alanine transaminase activity were higher in the control siRNA-transfected ob/ob mice than in the control siRNA-transfected Lean mice, but these parameters were reduced by GRK2 siRNA transfection into the ob/ob mice. GRK2 expression in epididymal fat was not altered among the 3 groups, although hepatic GRK2 expression was higher in the control siRNA-transfected ob/ob mice than in the control siRNA-transfected Lean mice. Additionally, we found that Akt interacted with GRK2 in the liver. Furthermore, phosphorylation levels of ERK1/2 and JNK were higher in the epididymal fats from the control siRNA-transfected ob/ob mice than in those from the control siRNA-transfected Lean mice, but they were lowered by transfection with GRK2 siRNA. The study results showed that GRK2 siRNA improved blood triglyceride levels and abnormal or excessive activity of mitogen-activated protein kinases in epididymal fat. This effect may be promoted by inhibition of the NEFA production pathway in the liver. Therefore, the interaction of organs (hepatic GRK2-epididymal fat) may help improve insulin resistance and diabetes-associated pathophysiology.
Collapse
|
121
|
Al-Sulaiti H, Diboun I, Banu S, Al-Emadi M, Amani P, Harvey TM, Dömling AS, Latiff A, Elrayess MA. Triglyceride profiling in adipose tissues from obese insulin sensitive, insulin resistant and type 2 diabetes mellitus individuals. J Transl Med 2018; 16:175. [PMID: 29940972 PMCID: PMC6019324 DOI: 10.1186/s12967-018-1548-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/15/2018] [Indexed: 01/01/2023] Open
Abstract
Background Lipid intermediates produced during triacylglycerols (TAGs) synthesis and lipolysis in adipocytes interfere with the intracellular insulin signaling pathway and development of insulin resistance. This study aims to compare TAG species and their fatty acid composition in adipose tissues from insulin sensitive (IS), insulin resistant (IR) and type 2 diabetes mellitus (T2DM) obese individuals. Methods Human subcutaneous and omental adipose tissue biopsies were obtained from 64 clinically characterized obese individuals during weight reduction surgery. TAGs were extracted from the adipose tissues using the Bligh and Dyer method, then were subjected to non-aqueous reverse phase ultra-high performance liquid chromatography and full scan mass spectrometry acquisition and data dependent MS/MS on LTQ dual cell linear ion trap. TAGs and their fatty acid contents were identified and compared between IS, IR and T2DM individuals and their levels were correlated with metabolic traits of participants and the adipogenic potential of preadipocyte cultures established from their adipose tissues. Results Data revealed 76 unique TAG species in adipose tissues identified based on their exact mass. Analysis of TAG levels revealed a number of TAGs that were significantly altered with disease progression including C46:4, C48:5, C48:4, C38:1, C50:3, C40:2, C56:3, C56:4, C56:7 and C58:7. Enrichment analysis revealed C12:0 fatty acid to be associated with TAGs least abundant in T2DM whereas C18:3 was found in both depleted and enriched TAGs in T2DM. Significant correlations of various adipose tissue-derived TAG species and metabolic traits were observed, including age and body mass index, systemic total cholesterol, TAGs, and interleukin-6 in addition to adipogenic potential of preadipocytes derived from the same adipose tissues. Conclusion Pilot data suggest that adipose tissues from obese IR and T2DM individuals exhibit TAG-specific signatures that may contribute to their increased risk compared to their IS counterparts. Future experiments are warranted to investigate the functional relevance of these specific lipidomic profiles. Electronic supplementary material The online version of this article (10.1186/s12967-018-1548-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Haya Al-Sulaiti
- Toxicology and Multipurpose Lab, Anti Doping Laboratory Qatar, Sports City, Doha, Qatar
| | - Ilhame Diboun
- Department of Economics, Mathematics and Statistics, Birkbeck, University of London, London, WC1E 7HX, UK
| | - Sameem Banu
- Toxicology and Multipurpose Lab, Anti Doping Laboratory Qatar, Sports City, Doha, Qatar
| | | | - Parvaneh Amani
- General Surgery Department, Al-Emdi Hospital, Doha, Qatar
| | - Thomas M Harvey
- Toxicology and Multipurpose Lab, Anti Doping Laboratory Qatar, Sports City, Doha, Qatar
| | - Alex S Dömling
- Department of Drug Design, University of Groningen, A. Deusinglaan 1, 9713 AV, Groningen, Netherlands
| | - Aishah Latiff
- Toxicology and Multipurpose Lab, Anti Doping Laboratory Qatar, Sports City, Doha, Qatar
| | - Mohamed A Elrayess
- Toxicology and Multipurpose Lab, Anti Doping Laboratory Qatar, Sports City, Doha, Qatar. .,Division of Medicine, Royal Free and University College Medical School, University College London, London, UK.
| |
Collapse
|
122
|
Guex E, Kouadio A, Fierz Y, Coti Bertrand P. Prise en charge diététique du sujet obèse à l’hôpital : quels régimes ? NUTR CLIN METAB 2018. [DOI: 10.1016/j.nupar.2018.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
123
|
Insulin resistance in obesity: an overview of fundamental alterations. Eat Weight Disord 2018; 23:149-157. [PMID: 29397563 DOI: 10.1007/s40519-018-0481-6] [Citation(s) in RCA: 208] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/12/2018] [Indexed: 12/14/2022] Open
Abstract
Obesity is a major health risk factor, and obesity-induced morbidity and complications account for huge costs for affected individuals, families, healthcare systems, and society at large. In particular, obesity is strongly associated with the development of insulin resistance, which in turn plays a key role in the pathogenesis of obesity-associated cardiometabolic complications, including metabolic syndrome components, type 2 diabetes, and cardiovascular diseases. Insulin sensitive tissues, including adipose tissue, skeletal muscle, and liver, are profoundly affected by obesity both at biomolecular and functional levels. Altered adipose organ function may play a fundamental pathogenetic role once fat accumulation has ensued. Modulation of insulin sensitivity appears to be, at least in part, related to changes in redox balance and oxidative stress as well as inflammation, with a relevant underlying role for mitochondrial dysfunction that may exacerbate these alterations. Nutrients and substrates as well as systems involved in host-nutrient interactions, including gut microbiota, have been also identified as modulators of metabolic pathways controlling insulin action. This review aims at providing an overview of these concepts and their potential inter-relationships in the development of insulin resistance, with particular regard to changes in adipose organ and skeletal muscle.
Collapse
|
124
|
An JY, Jheng HF, Nagai H, Sanada K, Takahashi H, Iwase M, Watanabe N, Kim YI, Teraminami A, Takahashi N, Nakata R, Inoue H, Seno S, Mastuda H, Kawada T, Goto T. A Phytol-Enriched Diet Activates PPAR-α in the Liver and Brown Adipose Tissue to Ameliorate Obesity-Induced Metabolic Abnormalities. Mol Nutr Food Res 2018; 62:e1700688. [DOI: 10.1002/mnfr.201700688] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 12/28/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Ji-Yeong An
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Huei-Fen Jheng
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Hiroyuki Nagai
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
- Gifu Prefectural Research Institute for Health and Environmental Science; Kakamigahara Japan
| | - Kohei Sanada
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Mari Iwase
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Natsumi Watanabe
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Young-Il Kim
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Aki Teraminami
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; The Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Uji Japan
| | - Rieko Nakata
- Department of Food Science and Nutrition; Nara Women's University; Nara Japan
| | - Hiroyasu Inoue
- Department of Food Science and Nutrition; Nara Women's University; Nara Japan
| | - Shigeto Seno
- Graduate School of Information Science and Technology; Osaka University; Osaka Japan
| | - Hideo Mastuda
- Graduate School of Information Science and Technology; Osaka University; Osaka Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; The Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Uji Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food; Division of Food Science and Biotechnology; Graduate School of Agriculture; Kyoto University; Uji Japan
- Research Unit for Physiological Chemistry; The Center for the Promotion of Interdisciplinary Education and Research; Kyoto University; Uji Japan
| |
Collapse
|
125
|
Li H, Wu G, Fang Q, Zhang M, Hui X, Sheng B, Wu L, Bao Y, Li P, Xu A, Jia W. Fibroblast growth factor 21 increases insulin sensitivity through specific expansion of subcutaneous fat. Nat Commun 2018; 9:272. [PMID: 29348470 PMCID: PMC5773530 DOI: 10.1038/s41467-017-02677-9] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 12/19/2017] [Indexed: 02/08/2023] Open
Abstract
Although the pharmacological effects of fibroblast growth factor 21 (FGF21) are well-documented, uncertainty about its role in regulating excessive energy intake remains. Here, we show that FGF21 improves systemic insulin sensitivity by promoting the healthy expansion of subcutaneous adipose tissue (SAT). Serum FGF21 levels positively correlate with the SAT area in insulin-sensitive obese individuals. FGF21 knockout mice (FGF21KO) show less SAT mass and are more insulin-resistant when fed a high-fat diet. Replenishment of recombinant FGF21 to a level equivalent to that in obesity restores SAT mass and reverses insulin resistance in FGF21KO, but not in adipose-specific βklotho knockout mice. Moreover, transplantation of SAT from wild-type to FGF21KO mice improves insulin sensitivity in the recipients. Mechanistically, circulating FGF21 upregulates adiponectin in SAT, accompanied by an increase of M2 macrophage polarization. We propose that elevated levels of endogenous FGF21 in obesity serve as a defense mechanism to protect against systemic insulin resistance. FGF21 has a number of beneficial metabolic effects. Here, Li et al. show that FGF21 promotes the healthy expansion of subcutaneous white adipose tissue, promoting the healthy expansion of fat tissue as a regulatory mechanism to maintain systemic insulin sensitivity during nutrient excess.
Collapse
Affiliation(s)
- Huating Li
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Guangyu Wu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qichen Fang
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Mingliang Zhang
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Xiaoyan Hui
- State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China.,Department of Medicine, The University of Hong Kong, Hong Kong, China
| | - Bin Sheng
- Department of Computer Science and Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Liang Wu
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Department of Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqian Bao
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Peng Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Hong Kong, China. .,Department of Medicine, The University of Hong Kong, Hong Kong, China.
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center of Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
126
|
Zabarsky G, Beek C, Hagman E, Pierpont B, Caprio S, Weiss R. Impact of Severe Obesity on Cardiovascular Risk Factors in Youth. J Pediatr 2018; 192:105-114. [PMID: 29246331 PMCID: PMC9338402 DOI: 10.1016/j.jpeds.2017.09.066] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 09/02/2017] [Accepted: 09/22/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To compare cardiovascular risk factor clustering (CVRFC) in severely obese youth with those with lower degrees of obesity. STUDY DESIGN We divided a childhood obesity clinic derived cohort into the degrees of obesity (class I, II, and III) and added a "class IV" category corresponding to >160% of the 95th centile of body mass index (BMI) for age and sex. In a cross-sectional analysis, we investigated the presence of CVRFC in 2244 participants; in 621 who were followed longitudinally, we investigated the determinants of endpoint CVRFC. RESULTS Class IV obesity was associated with increased risk for CVRFC compared with overweight (OR = 17.26, P < .001) at a similar magnitude to class III obesity (OR = 17.26, P < .001). Male children were at greater risk for presence of CVRFC (OR = 1.57, P = .03) compared with female children. Adiponectin (OR = 0.90, P < .001) and leptin levels (OR = 0.98, P = .008) were protective, independent of degree of obesity. Baseline class IV obesity was associated with increased risk compared with overweight of having CVRFC at follow-up (OR = 5.76, P = .001), to a similar extent as class III obesity (OR = 5.36, P = .001). Changes in the degree of obesity were significant predictors of CVRFC on follow-up (OR = 1.04, P < .01 per percent BMI change). CONCLUSIONS The metabolic risk associated with obesity in childhood is conferred prior to reaching class IV obesity. An individualized risk stratification approach in children with severe obesity should be based on presence of complications rather than simple BMI cutoffs. TRIAL REGISTRATION ClinicalTrials.gov NCT01967849.
Collapse
Affiliation(s)
- Gali Zabarsky
- Department of Human Metabolism and Nutrition, Braun School of Public Health, Hebrew University, Jerusalem, Israel
| | - Cherise Beek
- Department of Human Metabolism and Nutrition, Braun School of Public Health, Hebrew University, Jerusalem, Israel
| | - Emilia Hagman
- Department of Human Metabolism and Nutrition, Braun School of Public Health, Hebrew University, Jerusalem, Israel
| | | | - Sonia Caprio
- Department of Pediatrics, Yale University, New-Haven, CT, USA
| | - Ram Weiss
- Department of Human Metabolism and Nutrition, Braun School of Public Health, Hebrew University, Jerusalem, Israel.
| |
Collapse
|
127
|
Abstract
Adipose tissue and liver are central tissues in whole body energy metabolism. Their composition, structure, and function can be noninvasively imaged using a variety of measurement techniques that provide a safe alternative to an invasive biopsy. Imaging of adipose tissue is focused on quantitating the distribution of adipose tissue in subcutaneous and intra-abdominal (visceral) adipose tissue depots. Also, detailed subdivisions of adipose tissue can be distinguished with modern imaging techniques. Adipose tissue (or adipocyte) accumulation or infiltration of other organs can also be imaged, with intramuscular adipose tissue a common example. Although liver fat content is now accurately imaged using standard magnetic resonance imaging (MRI) techniques, inflammation and fibrosis are more difficult to determine noninvasively. Liver imaging efforts are therefore concerted on developing accurate imaging markers of liver fibrosis and inflammatory status. Magnetic resonance elastography (MRE) is presently the most reliable imaging technique for measuring liver fibrosis but requires an external device for introduction of shear waves to the liver. Methods using multiparametric diffusion, perfusion, relaxometry, and hepatocyte-specific MRI contrast agents may prove to be more easily implemented by clinicians, provided they reach similar accuracy as MRE. Adipose tissue imaging is experiencing a revolution with renewed interest in characterizing and identifying distinct adipose depots, among them brown adipose tissue. Magnetic resonance spectroscopy provides an interesting yet underutilized way of imaging adipose tissue metabolism through its fatty acid composition. Further studies may shed light on the role of fatty acid composition in different depots and why saturated fat in subcutaneous adipose tissue is a marker of high insulin sensitivity.
Collapse
Affiliation(s)
- Jesper Lundbom
- Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University, Düsseldorf, Germany
- German Center for Diabetes Research, München-Neuherberg, Düsseldorf, Germany
- HUS Medical Imaging Center, Radiology, Helsinki University Central Hospital, University of Helsinki, Finland
| |
Collapse
|
128
|
Yoshino S, Awa R, Miyake Y, Fukuhara I, Sato H, Ashino T, Tomita S, Kuwahara H. Daily intake of Kaempferia parviflora extract decreases abdominal fat in overweight and preobese subjects: a randomized, double-blind, placebo-controlled clinical study. Diabetes Metab Syndr Obes 2018; 11:447-458. [PMID: 30214264 PMCID: PMC6120512 DOI: 10.2147/dmso.s169925] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
PURPOSE Obesity is a serious problem, which is now a worldwide health problem. Kaempferia parviflora extract (KPE) exhibits anti-obesity effects in animals. However, as no clinical trials have evaluated the anti-obesity effects of KPE in humans, we examined the effects of KPE in reducing abdominal fat in overweight and preobese Japanese subjects. MATERIALS AND METHODS A 12-week, single-center, randomized, double-blind, placebo-controlled clinical trial was conducted. Seventy-six subjects (males and females aged 20 to <65 years) with a body mass index ≥24 and <30 kg/m2 were randomly assigned into two groups. The subjects in each group ingested one capsule of placebo or active KPE (containing 150 mg of KPE) once daily for 12 weeks. The primary outcome was reduction in visceral fat area as determined by computed tomography scanning. The key secondary outcomes were reductions in subcutaneous fat area and total fat area. Subgroup analysis was also performed in healthy subjects without dyslipidemia, hypertension, or hyperglycemia. The safety of KPE ingestion was also evaluated. RESULTS Compared with the placebo group, the active KPE group exhibited significant reduction in abdominal fat area (visceral, subcutaneous, and total fat) and triglyceride levels after 12 weeks. Subgroup analyses demonstrated a significant reduction in abdominal fat area and triglyceride levels in healthy subjects compared with the placebo group after 12 weeks. Neither group exhibited adverse events related to the test foods or clinically relevant abnormal changes in physical, biochemical, or hematologic parameters, or in urinalysis results and medical interview. CONCLUSION Daily ingestion of KPE safely reduces body fat, particularly abdominal fat, in Japanese overweight and preobese subjects.
Collapse
Affiliation(s)
- Susumu Yoshino
- Research and Development Division, Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan,
| | - Riyo Awa
- Research and Development Division, Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan,
| | - Yasuo Miyake
- Research and Development Division, Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan,
| | | | - Hisao Sato
- Clinical Research Department, New Drug Research Center, Inc., Hokkaido, Japan
| | - Toyotada Ashino
- Clinical Research Department, New Drug Research Center, Inc., Hokkaido, Japan
| | - Shinpei Tomita
- Clinical Research Department, New Drug Research Center, Inc., Hokkaido, Japan
| | - Hiroshige Kuwahara
- Research and Development Division, Research Center, Maruzen Pharmaceuticals Co., Ltd., Hiroshima, Japan,
| |
Collapse
|
129
|
Fernández-Alfonso MS, Somoza B, Tsvetkov D, Kuczmanski A, Dashwood M, Gil-Ortega M. Role of Perivascular Adipose Tissue in Health and Disease. Compr Physiol 2017; 8:23-59. [PMID: 29357124 DOI: 10.1002/cphy.c170004] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Perivascular adipose tissue (PVAT) is cushion of fat tissue surrounding blood vessels, which is phenotypically different from other adipose tissue depots. PVAT is composed of adipocytes and stromal vascular fraction, constituted by different populations of immune cells, endothelial cells, and adipose-derived stromal cells. It expresses and releases an important number of vasoactive factors with paracrine effects on vascular structure and function. In healthy individuals, these factors elicit a net anticontractile and anti-inflammatory paracrine effect aimed at meeting hemodynamic and metabolic demands of specific organs and regions of the body. Pathophysiological situations, such as obesity, diabetes or hypertension, induce changes in its amount and in the expression pattern of vasoactive factors leading to a PVAT dysfunction in which the beneficial paracrine influence of PVAT is shifted to a pro-oxidant, proinflammatory, contractile, and trophic environment leading to functional and structural cardiovascular alterations and cardiovascular disease. Many different PVATs surrounding a variety of blood vessels have been described and exhibit regional differences. Both protective and deleterious influence of PVAT differs regionally depending on the specific vascular bed contributing to variations in the susceptibility of arteries and veins to vascular disease. PVAT therefore, might represent a novel target for pharmacological intervention in cardiovascular disease. © 2018 American Physiological Society. Compr Physiol 8:23-59, 2018.
Collapse
Affiliation(s)
| | - Beatriz Somoza
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| | - Dmitry Tsvetkov
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, Department of Pharmacology and Experimental Therapy, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research, Tübingen, Germany
| | - Artur Kuczmanski
- Department of Anestesiology, Perioperative and Pain Medicine, HELIOS Klinikum, Berlin-Buch GmbH, Germany
| | - Mick Dashwood
- Royal Free Hospital Campus, University College Medical School, London, United Kingdom
| | - Marta Gil-Ortega
- Departamento de Ciencias Farmacéuticas y de la Salud, Facultad de Farmacia, Universidad CEU-San Pablo, Madrid, Spain
| |
Collapse
|
130
|
Cai X, Hayashi S, Fang C, Hao S, Wang X, Nishiguchi S, Tsutsui H, Sheng J. Pu'erh tea extract-mediated protection against hepatosteatosis and insulin resistance in mice with diet-induced obesity is associated with the induction of de novo lipogenesis in visceral adipose tissue. J Gastroenterol 2017; 52:1240-1251. [PMID: 28364190 DOI: 10.1007/s00535-017-1332-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 03/16/2017] [Indexed: 02/04/2023]
Abstract
BACKGROUND White adipose tissue (WAT) is important for the maintenance of metabolic homeostasis, and metabolic syndrome is sometimes associated with WAT dysfunction in humans and animals. WAT reportedly plays a key, beneficial role in the maintenance of glucose and lipid homeostasis during de novo lipogenesis (DNL). Pu'erh tea extract (PTE) can inhibit harmful, ectopic DNL in the liver, thus protecting against hepatosteatosis, in mice with diet-induced obesity. We examined whether PTE could induce DNL in WAT and consequently protect against hepatosteatosis. METHODS C57BL/6 male mice were fed a high-fat diet (HFD) with/without PTE for 16 weeks. Systemic insulin sensitivity was determined using HOMA-IR, insulin- and glucose-tolerance tests, and WAT adipogenesis was evaluated by histological analysis. Adipogenesis-, inflammation-, and DNL-related gene expression in visceral AT (VAT) and subcutaneous AT (SAT) was measured using quantitative reverse transcription-PCR. Regression analysis was used to investigate the association between DNL in WAT and systemic insulin resistance or hepatosteatosis. RESULTS Pu'erh tea extract significantly reduced the gain of body weight and SAT, but not VAT adiposity, in mice fed the high-fat diet and induced adipogenesis in VAT. The expression of DNL-related genes, including Glut4, encoding an important insulin-regulated glucose transporter (GLUT4), were highly elevated in VAT. Moreover, PTE inhibited VAT inflammation by simultaneously downregulating inflammatory molecules and inducing expression of Gpr120 that encodes an anti-inflammatory and pro-adipogenesis receptor (GPR-120) that recognizes unsaturated long-chain fatty acids, including DNL products. The expression of DNL-related genes in VAT was inversely correlated with hepatosteatosis and systemic insulin resistance. CONCLUSIONS Activation of DNL in VAT may explain PTE-mediated alleviation of hepatosteatosis symptoms and systemic insulin resistance.
Collapse
Affiliation(s)
- Xianbin Cai
- Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan
| | - Shuhei Hayashi
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
- Department of Pu-erh Tea and Medical Science, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Chongye Fang
- Department of Pu-erh Tea and Medical Science, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
- Key Laboratory of Pu-erh Tea Science, The Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
| | | | - Xuanjun Wang
- Key Laboratory of Pu-erh Tea Science, The Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, China
| | - Shuhei Nishiguchi
- Department of Internal Medicine, Hyogo College of Medicine, 1-1 Mukogawa-cho, Nishinomiya, Hyogo, 663-8501, Japan.
| | - Hiroko Tsutsui
- Department of Microbiology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
- Department of Pu-erh Tea and Medical Science, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, The Ministry of Education, Yunnan Agricultural University, Kunming, 650201, China.
- State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Kunming, China.
- Pu'erh Tea Research Institute, Pu'erh, China.
| |
Collapse
|
131
|
Matsushita K, Dzau VJ. Mesenchymal stem cells in obesity: insights for translational applications. J Transl Med 2017; 97:1158-1166. [PMID: 28414326 DOI: 10.1038/labinvest.2017.42] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 02/24/2017] [Indexed: 12/11/2022] Open
Abstract
Obesity is now a major public health problem worldwide. Lifestyle modification to reduce the characteristic excess body adiposity is important in the treatment of obesity, but effective therapeutic intervention is still needed to control what has become an obesity epidemic. Unfortunately, many anti-obesity drugs have been withdrawn from market due to adverse side effects. Bariatric surgery therefore remains the most effective therapy for severe cases, although such surgery is invasive and researchers continue to seek new control strategies for obesity. Mesenchymal stem cells (MSCs) are a major source of adipocyte generation, and studies have been conducted into the potential roles of MSCs in treating obesity. However, despite significant progress in stem cell research and its potential applications for obesity, adipogenesis is a highly complex process and the molecular mechanisms governing MSC adipogenesis remain ill defined. In particular, successful clinical application of MSCs will require extensive identification and characterization of the transcriptional regulators controlling MSC adipogenesis. Since obesity is associated with the incidence of multiple important comorbidities, an in-depth understanding of the relationship between MSC adipogenesis and the comorbidities of obesity is also necessary to evaluate the potential of effective and safe MSC-based therapies for obesity. In addition, brown adipogenesis is an attractive topic from the viewpoint of therapeutic innovation and future research into MSC-based brown adipogenesis could lead to a novel breakthrough. Ongoing stem cell studies and emerging research fields such as epigenetics are expected to elucidate the complicated mechanisms at play in MSC adipogenesis and develop novel MSC-based therapeutic options for obesity. This review discusses the current understanding of MSCs in adipogenesis and their potential clinical applications for obesity.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Division of Cardiology, Second Department of Internal Medicine, Kyorin University School of Medicine, Tokyo, Japan
| | | |
Collapse
|
132
|
Dossus L, Rinaldi S, Biessy C, Hernandez M, Lajous M, Monge A, Ortiz-Panozo E, Yunes E, Lopez-Ridaura R, Torres-Mejía G, Romieu I. Circulating leptin and adiponectin, and breast density in premenopausal Mexican women: the Mexican Teachers' Cohort. Cancer Causes Control 2017; 28:939-946. [PMID: 28677026 DOI: 10.1007/s10552-017-0917-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/21/2017] [Indexed: 01/01/2023]
Abstract
PURPOSE Leptin and adiponectin are produced by the adipose tissue. Mammographic density (MD) is one of the strongest predictors of breast cancer (BC) and is highly influenced by adiposity. How the interplay between MD, obesity, and obesity-related biomarkers influences BC risk, however, is still unknown, especially in premenopausal women, where adiposity seems to be protective for BC. The aim of the present study was to explore the association between circulating leptin, adiponectin, and their ratio, with MD in Mexican premenopausal women who are part of the large Mexican Teachers' Cohort (MTC). METHODS A subsample of 2,084 women from the MTC participated in a clinical evaluation. Of them, 574 premenopausal women were randomly selected, from four MD strata. Serum leptin and adiponectin concentrations were measured by immunoassays. Multivariate regression analyses were performed to compare means of MD by quartiles of adipokines and their ratio. RESULTS High leptin and leptin/adiponectin ratio levels were significantly associated with lower percentage MD and higher absolute and non-absolute dense tissue areas. High adiponectin levels were significantly associated with lower absolute dense and non-dense tissue areas, but not with percentage MD. After adjustment for BMI, only the associations between percentage MD and absolute non-dense tissue area with leptin remained statistically significant. CONCLUSIONS Leptin, adiponectin, and their ratio were associated with MD; however, only the positive association with leptin seemed to be independent from overall obesity.
Collapse
Affiliation(s)
- L Dossus
- International Agency for Research on Cancer [IARC], Lyon, France
| | - S Rinaldi
- International Agency for Research on Cancer [IARC], Lyon, France
| | - C Biessy
- International Agency for Research on Cancer [IARC], Lyon, France
| | - M Hernandez
- International Agency for Research on Cancer [IARC], Lyon, France
| | - M Lajous
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico.
- Department of Epidemiology, Harvard School of Public Health, Boston, MA, USA.
| | - A Monge
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| | - E Ortiz-Panozo
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| | - E Yunes
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| | - R Lopez-Ridaura
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| | - G Torres-Mejía
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| | - I Romieu
- International Agency for Research on Cancer [IARC], Lyon, France
- Center for Research on Population Health, National Institute of Public Health, 7ª Cerrada Fray Pedro de Gante # 50, Mexico, 14000, Mexico
| |
Collapse
|
133
|
Bischoff SC, Boirie Y, Cederholm T, Chourdakis M, Cuerda C, Delzenne NM, Deutz NE, Fouque D, Genton L, Gil C, Koletzko B, Leon-Sanz M, Shamir R, Singer J, Singer P, Stroebele-Benschop N, Thorell A, Weimann A, Barazzoni R. Towards a multidisciplinary approach to understand and manage obesity and related diseases. Clin Nutr 2017; 36:917-938. [DOI: 10.1016/j.clnu.2016.11.007] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/03/2016] [Accepted: 11/03/2016] [Indexed: 12/13/2022]
|
134
|
Keating SE, Hackett DA, Parker HM, Way KL, O'Connor HT, Sainsbury A, Baker MK, Chuter VH, Caterson ID, George J, Johnson NA. Effect of resistance training on liver fat and visceral adiposity in adults with obesity: A randomized controlled trial. Hepatol Res 2017; 47:622-631. [PMID: 27480242 DOI: 10.1111/hepr.12781] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 07/23/2016] [Accepted: 07/26/2016] [Indexed: 02/08/2023]
Abstract
AIM Regular aerobic exercise reduces visceral adipose tissue (VAT) and liver fat, however, not all individuals are able to adopt and adhere to such programs. Progressive resistance training (PRT) may be an alternative therapy, but there is limited available evidence. We examined the efficacy of PRT as per current exercise guidelines, compared with sham exercise placebo on liver fat and VAT. METHODS Twenty-nine inactive and overweight/obese (body mass index ≥25 kg/m2 ) adults (age 29-59) were randomized to receive 8 weeks of PRT (n = 15, 10 exercises per session, 8-12 repetitions, 2-3 sets per exercise at 80-85% of one-repetition maximum, 3 days per week) or a sham exercise placebo control (CON) (n = 14). Change in liver fat, VAT, and abdominal s.c. adipose tissue (SAT) were assessed by magnetic resonance spectroscopy and imaging). RESULTS There were no significant group by time interactions for change in liver fat in PRT versus CON groups (-0.07 ± 0.31% vs. 0.55 ± 0.77%, respectively, P = 0.19), VAT (-175 ± 85 cm3 vs. 10 ± 64 cm3 , respectively, P = 0.11), or abdominal SAT (-436 ± 245 cm3 vs. 127.29 ± 182 cm3 , respectively, P = 0.10) despite a significant increase in muscle volume (55 ± 78 cm3 vs. -0.04 ± 8 cm3 , respectively, P = 0.03). CONCLUSION Traditional PRT is not effective for reducing liver fat in overweight/obese adults compared with placebo control. Although PRT has known metabolic benefits, an adequate volume of aerobic exercise should be promoted if liver fat is the therapeutic target.
Collapse
Affiliation(s)
- Shelley E Keating
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia.,Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Daniel A Hackett
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia
| | - Helen M Parker
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia
| | - Kimberley L Way
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia
| | - Helen T O'Connor
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia
| | - Amanda Sainsbury
- Boden Institute of Obesity, Nutrition, Exercise and Eating Disorders, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Michael K Baker
- School of Exercise Science, Australian Catholic University, Sydney, New South Wales, Australia
| | - Vivienne H Chuter
- School of Health Sciences, University of Newcastle, Newcastle, New South Wales, Australia
| | - Ian D Caterson
- Boden Institute of Obesity, Nutrition, Exercise and Eating Disorders, Sydney Medical School, University of Sydney, Sydney, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Millennium Institute and Westmead Hospital, University of Sydney, Sydney, Australia
| | - Nathan A Johnson
- Discipline of Exercise and Sport Science, University of Sydney, Sydney, Australia.,Boden Institute of Obesity, Nutrition, Exercise and Eating Disorders, Sydney Medical School, University of Sydney, Sydney, Australia
| |
Collapse
|
135
|
Wan Y, Bao X, Huang J, Zhang X, Liu W, Cui Q, Jiang D, Wang Z, Liu R, Wang Q. Novel GLP-1 Analog Supaglutide Reduces HFD-Induced Obesity Associated with Increased Ucp-1 in White Adipose Tissue in Mice. Front Physiol 2017; 8:294. [PMID: 28555111 PMCID: PMC5430033 DOI: 10.3389/fphys.2017.00294] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/24/2017] [Indexed: 12/14/2022] Open
Abstract
GLP-1, an important incretin hormone plays an important role in the regulation of glucose homeostasis. However, the therapeutic use of native GLP-1 is limited due to its short half-life. We recently developed a novel GLP-1 mimetics (supaglutide) by genetically engineering recombinant fusion protein production techniques. We demonstrated that this formulation possessed long-lasting GLP-1 actions and was effective in glycemic control in both type 1 and type 2 diabetes rodent models. Here, we investigated the effects of supaglutide in regulating energy homeostasis in obese mice. Mice were fed with high-fat diet (HFD) for 6 months to induce obesity and then subjected to supaglutide treatment (300 μg/kg, bi-weekly for 4 weeks), and placebo as control. Metabolic conditions were monitored and energy expenditure was assessed by indirect calorimetry (CLAMS). Cold tolerance test was performed to evaluate brown-adipose tissue (BAT) activities in response to cold challenge. Glucose tolerance and insulin resistance were evaluated by intraperitoneal glucose tolerance test and insulin tolerance tests. Liver and adipose tissues were collected for histology analysis. Expression of uncoupling protein 1(Ucp1) in adipose tissues was evaluated by Western blotting. We found that supaglutide treatment reduced body weight, which was associated with reduced food intake. Compared to the placebo control, supaglutide treatment improved lipid profile, i.e., significantly decreased circulating total cholesterol levels, declined serum triglyceride, and free fatty acid levels. Importantly, the intervention significantly reduced fatty liver, decreased liver triglyceride content, and concomitantly ameliorated liver injury exemplified by declined hepatic alanine aminotransferase (ALT) and aspartic transaminase (AST) content. Remarkably, supaglutide reduced hepatic lipid accumulation and altered morphometry in favor of small adipocytes in fat. This is consistent with the observation that supaglutide increased tolerance of the mice to cold environment associated with up-regulation of Ucp1 in the inguinal fat. Furthermore, supaglutide improved glucose tolerance, and insulin sensitivity in the obese mice suggesting improved glucose and energy homeostasis. Our findings suggest that supaglutide exerts beneficial effect on established obesity through reducing energy intake and is associated with brown remodeling of white adipose tissue.
Collapse
Affiliation(s)
- Yun Wan
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China
| | - Xi Bao
- Yinnuo Pharmaceutical Technology Co. Ltd.Shanghai, China
| | - Jiabao Huang
- Yinnuo Pharmaceutical Technology Co. Ltd.Shanghai, China
| | - Xiangyu Zhang
- Yinnuo Pharmaceutical Technology Co. Ltd.Shanghai, China
| | - Wenjuan Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China.,Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's HospitalToronto, ON, Canada
| | - Qiaoli Cui
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China
| | - Dongdong Jiang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China
| | - Zhihong Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China
| | - Rui Liu
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Fudan UniversityShanghai, China.,Yinnuo Pharmaceutical Technology Co. Ltd.Shanghai, China.,Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's HospitalToronto, ON, Canada.,Departments of Physiology and Medicine, Faculty of Medicine, University of TorontoON, Canada
| |
Collapse
|
136
|
Lewitt MS. The Role of the Growth Hormone/Insulin-Like Growth Factor System in Visceral Adiposity. BIOCHEMISTRY INSIGHTS 2017; 10:1178626417703995. [PMID: 28469442 PMCID: PMC5404904 DOI: 10.1177/1178626417703995] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 03/19/2017] [Indexed: 12/18/2022]
Abstract
There is substantial evidence that the growth hormone (GH)/insulin-like growth factor (IGF) system is involved in the pathophysiology of obesity. Both GH and IGF-I have direct effects on adipocyte proliferation and differentiation, and this system is involved in the cross-talk between adipose tissue, liver, and pituitary. Transgenic animal models have been of importance in identifying mechanisms underlying these interactions. It emerges that this system has key roles in visceral adiposity, and there is a rationale for targeting this system in the treatment of visceral obesity associated with GH deficiency, metabolic syndrome, and lipodystrophies. This evidence is reviewed, gaps in knowledge are highlighted, and recommendations are made for future research.
Collapse
Affiliation(s)
- Moira S Lewitt
- School of Health, Nursing & Midwifery, University of the West of Scotland, Paisley, UK
| |
Collapse
|
137
|
Xia Y, Fu Y, Wang Y, Qian Y, Li X, Xu H, Zou J, Guan J, Yi H, Meng L, Tang X, Zhu H, Yu D, Zhou H, Su K, Yin S. Prevalence and Predictors of Atherogenic Serum Lipoprotein Dyslipidemia in Women with Obstructive Sleep Apnea. Sci Rep 2017; 7:41687. [PMID: 28134311 PMCID: PMC5278373 DOI: 10.1038/srep41687] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/21/2016] [Indexed: 12/19/2022] Open
Abstract
Obstructive sleep apnea (OSA) is associated with dyslipidemia. However, no study has focused on dyslipidemia in women with OSA. The aim of this study was to determine the prevalence and risk factors for dyslipidemia in women with OSA. Between 2007 and 2013, 570 eligible female patients with suspected OSA were consecutively recruited. The analyzed data consisted of polysomnography parameters, biochemical indicators, and anthropometric measurements. Serum lipid levels and dyslipidemia were compared. Binary logistic regression and multivariate linear regression models were used to determine the independent risk factors influencing serum lipids. After multivariate adjustment, there were essentially no major differences in serum lipid levels among patients with no to mild, moderate, and severe OSA nor did serum lipid levels change with OSA severity. Dyslipidemia in total cholesterol, triglycerides, low-density lipoprotein cholesterol, apolipoproteins(apo) B and apoE increased with OSA severity, but only in non-obese subjects and those <55 years of age. Age, body mass index, waist to hip ratio, glucose and insulin were major risk factors for most serum lipids after multivariate adjustments. Our results indicate that, in women with OSA, age, obesity/central obesity, and insulin resistance are major determinants of dyslipidemia.
Collapse
Affiliation(s)
- Yunyan Xia
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Yiqun Fu
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Yuyu Wang
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Yingjun Qian
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Xinyi Li
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Huajun Xu
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Jianyin Zou
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Jian Guan
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Hongliang Yi
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Lili Meng
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Xulan Tang
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Huaming Zhu
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Dongzhen Yu
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Huiqun Zhou
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Kaiming Su
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| | - Shankai Yin
- Department of Otolaryngology Head and Neck Surgery & Center of Sleep Medicine, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Yishan Road 600, Shanghai, 200233, China
- Otolaryngological Institute of Shanghai Jiao Tong University, Yishan Road 600, Shanghai, 200233, China
- Clinical Research Center, Shanghai Jiao Tong University School of Medicine, South Chongqing Road 225, Shanghai, 200020, China
| |
Collapse
|
138
|
Daan NMP, Muka T, Koster MPH, Roeters van Lennep JE, Lambalk CB, Laven JSE, Fauser CGKM, Meun C, de Rijke YB, Boersma E, Franco OH, Kavousi M, Fauser BCJM. Cardiovascular Risk in Women With Premature Ovarian Insufficiency Compared to Premenopausal Women at Middle Age. J Clin Endocrinol Metab 2016; 101:3306-15. [PMID: 27300572 DOI: 10.1210/jc.2016-1141] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT A young age at menopause has been associated with increased cardiovascular disease (CVD) risk. OBJECTIVE To compare the cardiovascular risk profile between women with premature ovarian insufficiency (POI) and premenopausal controls of comparable age. DESIGN Cross-sectional case control study. SETTING Two university medical centers. PARTICIPANTS Women above 45 years of age who were previously diagnosed with POI (n = 83) and premenopausal population controls of comparable age (n = 266). MAIN OUTCOME MEASURES Blood pressure, body mass index, waist circumference, electrocardiogram, bilateral carotid intima media thickness, estradiol, T, androstenedione, dehydroepiandrosterone sulfate, SHBG, insulin, glucose, lipids, TSH, free T4, N-terminal pro-B-type natriuretic peptide, C-reactive protein, uric acid, creatinine, and homocysteine were measured. Potential associations between POI status and subclinical atherosclerosis were assessed. RESULTS Women with POI exhibited an increased waist circumference (β = 5.7; 95% confidence interval [CI], 1.6, 9.9), C-reactive protein (β = 0.75; 95% CI, 0.43, 1.08), free T4 levels (β = 1.5; 95% CI, 0.6, 2.4), and lower N-terminal pro-B-type natriuretic peptide (β = -0.35; 95% CI, -0.62, -0.08), estradiol (β = -1.98; 95% CI, -2.48, -1.48), T (β = -0.21; 95% CI, -0.37, -0.06), and androstenedione (β = -0.54; 95% CI, -0.71, -0.38) concentrations compared to controls, after adjusting for confounders. After adjustment, a trend toward increased hypertension (odds ratio = 2.1; 95% CI, 0.99; 4.56) and decreased kidney function was observed in women with POI (creatinine β = 3.5; 95% CI, -0.05, 7.1; glomerular filtration rate β = -3.5; 95% CI, -7.5, 0.46). Women with POI exhibited a lower mean carotid intima media thickness (β = -0.17; 95% CI, -0.21, -0.13) and decreased odds of plaque presence compared to controls (odds ratio = 0.08; 95% CI, 0.03; 0.26). CONCLUSIONS Women with POI exhibited an unfavorable cardiovascular risk profile, including higher abdominal fat, elevated chronic inflammatory factors, and a trend toward increased hypertension and impaired kidney function compared to controls. However, we observed no signs of increased subclinical atherosclerosis in women with POI. Additional studies are required to identify specific determinants of long-term CVD risk in women with POI.
Collapse
Affiliation(s)
- Nadine M P Daan
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Taulant Muka
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Maria P H Koster
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Jaenine E Roeters van Lennep
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Cornelis B Lambalk
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Joop S E Laven
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Clemens G K M Fauser
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Cindy Meun
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Yolanda B de Rijke
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Eric Boersma
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Oscar H Franco
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Maryam Kavousi
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| | - Bart C J M Fauser
- Department of Reproductive Medicine and Gynecology (N.M.P.D., M.P.H.K., B.C.J.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Epidemiology (T.M., O.H.F., M.K.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Internal Medicine (J.E.R.v.L.), Division Vascular Medicine, Erasmus Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Obstetrics and Gynecology (C.B.L.), VU University Medical Center Amsterdam, 1081 HV Amsterdam, The Netherlands; Department of Obstetrics and Gynecology (J.S.E.L., C.M.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; Department of Cardiology (C.G.K.M.F.), University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands; Department of Clinical Chemistry (Y.B.d.R.), Erasmus MC, University Medical Center Rotterdam, 3015 CE Rotterdam, The Netherlands; and Department of Cardiology (E.B.), Erasmus MC, University Medical Center Rotterdam, Cardiovascular Research School Erasmus University Rotterdam, 3015 CE Rotterdam, The Netherlands
| |
Collapse
|
139
|
Park SK, Peng Q, Bielak LF, Silver KD, Peyser PA, Mitchell BD. Arsenic exposure is associated with diminished insulin sensitivity in non-diabetic Amish adults. Diabetes Metab Res Rev 2016; 32:565-71. [PMID: 26663816 PMCID: PMC4995145 DOI: 10.1002/dmrr.2769] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 10/30/2015] [Accepted: 11/26/2015] [Indexed: 01/01/2023]
Abstract
BACKGROUND Substantial evidence supports an association between diabetes and arsenic at high exposure levels, but results are mixed at low exposure levels. The aetiology of diabetes involves insulin resistance and β-cell dysfunction. However, only a few epidemiologic studies have examined measures of insulin resistance and β-cell function in relation to arsenic exposure, and no studies have tested for associations with the oral glucose tolerance test (OGTT). We examined the association between urinary total arsenic and OGTT-based markers of insulin sensitivity and β-cell function. METHODS We studied 221 non-diabetic adults (mean age = 52.5 years) from the Amish Family Diabetes Study. We computed OGTT-based validated measures of insulin sensitivity and β-cell function. Generalized estimating equations accounting for sibship were used to estimate associations. RESULTS After adjusting for age, sex, waist-to-hip ratio and urinary creatinine, an interquartile range increase in urinary total arsenic (6.24 µg/L) was significantly, inversely associated with two insulin sensitivity measures (Stumvoll metabolic clearance rate = -0.23 mg/(kg min), (95% CI: -0.38, -0.089), p = 0.0015; Stumvoll insulin sensitivity index = -0.0029 µmol/(kg min pM), (95% CI: -0.0047, -0.0011), p = 0.0015). Urinary total arsenic was also significantly associated with higher fasting glucose levels (0.57 mg/dL (95% CI: 0.06, 1.09) per interquartile range increase, p = 0.029). No significant associations were found between urinary total arsenic and β-cell function measures. CONCLUSIONS This preliminary study found that urinary total arsenic was associated with insulin sensitivity but not β-cell function measures, suggesting that low-level arsenic exposure may influence diabetes risk through impairing insulin sensitivity. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Sung Kyun Park
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
- Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Qing Peng
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Kristi D. Silver
- Departments of Medicine and Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD
| | - Patricia A. Peyser
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Braxton D. Mitchell
- Departments of Medicine and Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD
- Department of Veterans Affairs and Veterans Affairs Medical Center Baltimore Geriatric Research Education and Clinical Center (GRECC)
| |
Collapse
|
140
|
Matsushita K, Wu Y, Pratt RE, Dzau VJ. Deletion of angiotensin II type 2 receptor accelerates adipogenesis in murine mesenchymal stem cells via Wnt10b/beta-catenin signaling. J Transl Med 2016; 96:909-17. [PMID: 27295344 PMCID: PMC4965305 DOI: 10.1038/labinvest.2016.66] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 04/15/2016] [Accepted: 05/03/2016] [Indexed: 12/17/2022] Open
Abstract
Recent evidence suggests that the renin-angiotensin system (RAS) has a vital role in adipocyte biology and the pathophysiology of metabolic syndrome. Obesity is the main culprit of metabolic syndrome; and mesenchymal stem cells (MSCs) have been forwarded as a major source of adipocyte generation. Previously, we reported that MSCs have a local RAS and that pharmacological blockade of angiotensin II type 2 receptor (AT2R) promotes adipogenesis in human MSCs. However, the definitive roles of AT2R and how AT2R functions in adipogenesis remains unknown. To this end, we employed AT2R-null murine MSCs to characterize how AT2R affects the differentiation of MSCs into adipocytes. Murine MSCs were isolated from AT2R-null mice and wild-type littermates, grown to confluency, and then differentiated into adipocytes. Adipogenesis was quantitated by assessing the lipid droplet accumulation. Using the lipophilic fluorescent dye, the AT2R-null cells showed significantly increased total fluorescence (261.6±49.6% vs littermate) on day 7. Oil red O staining followed by extraction of the absorbed dye and measurement of the absorbance on day 14 also exhibited significantly increased lipid droplet accumulation in the AT2R-null cells (202.7±14.1% vs littermate). We also examined the expression of adipogenic marker genes by quantitative RT-PCR. The AT2R-null group exhibited significantly increased expression of PPAR-gamma, fatty acid synthase, and adiponectin (vs littermate). We further examined the role of Wnt10b/beta-catenin signaling, which reportedly has an important inhibitory role in adipogenesis. The AT2R-null group exhibited significantly decreased Wnt10b expression accompanied by decreased beta-catenin (vs littermate). Our results thus revealed that the AT2R inhibits adipogenic differentiation in murine MSCs. Moreover, this inhibitory effect is associated with Wnt10b/beta-catenin signaling. These results provide important insights into the pathophysiology of obesity and obesity-related consequences such as metabolic syndrome, hinting at possible future therapies.
Collapse
Affiliation(s)
- Kenichi Matsushita
- Division of Cardiology, Department of Medicine, Duke University Medical Center, GSRB II Bldg., Durham, NC 27710, USA, Division of Cardiology, Second Department of Internal Medicine, Kyorin University School of Medicine, Tokyo 181-8611, Japan
| | - Yaojiong Wu
- Division of Cardiology, Department of Medicine, Duke University Medical Center, GSRB II Bldg., Durham, NC 27710, USA
| | - Richard E Pratt
- Division of Cardiology, Department of Medicine, Duke University Medical Center, GSRB II Bldg., Durham, NC 27710, USA
| | - Victor J Dzau
- Division of Cardiology, Department of Medicine, Duke University Medical Center, GSRB II Bldg., Durham, NC 27710, USA, National Academy of Medicine, 500 Fifth St NW, Washington, DC 20001, USA
| |
Collapse
|
141
|
Wang X, You T, Murphy K, Lyles MF, Nicklas BJ. Addition of Exercise Increases Plasma Adiponectin and Release from Adipose Tissue. Med Sci Sports Exerc 2016; 47:2450-5. [PMID: 25811948 DOI: 10.1249/mss.0000000000000670] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Adiponectin is an adipose tissue-derived anti-inflammatory protein that is down-regulated in obesity. The effects of caloric restriction and exercise-induced weight loss on adiponectin are not clear. PURPOSE To determine whether addition of aerobic exercise training to caloric restriction has additive effects over caloric restriction alone on circulating adiponectin concentrations and adiponectin release from abdominal and gluteal adipose tissue. METHODS Overweight or obese (body mass index, 25-40 kg·m(-2); waist >88 cm) postmenopausal women were randomized to 20-wk caloric restriction with and without aerobic exercise (CR + EX, n = 48; and CR, n = 22). Blood samples were collected for measuring plasma adiponectin concentration, and abdominal and gluteal subcutaneous adipose tissue biopsies were performed in a subgroup to determine in vitro adiponectin release, before and after the interventions. RESULTS The interventions elicited similar amounts of weight loss (CR + EX, -11.3 ± 4.6 kg; CR,-11.2 ± 3.4 kg) and fat loss (CR + EX, -8.0 ± 3.5 kg; CR, -7.4 ± 2.7 kg). The two groups had differential changes in plasma adiponectin concentrations (for interaction, P = 0.014); CR + EX increased (6.9 ± 3.9 to 8.5 ± 4.9 μg·mL(-1); P = 0.0001), whereas CR did not alter (6.4 ± 4.4 to 6.5 ± 4.5 μg·mL(-1); P = 0.42) plasma adiponectin. Likewise, adiponectin release from abdominal and gluteal subcutaneous adipose tissue increased with CR + EX (P = 0.0076 and P = 0.089, respectively) but did not change with CR (P = 0.13 and P = 0.95, respectively). CONCLUSION Despite similar reductions in body weight and fat mass, the addition of aerobic exercise to caloric restriction increased plasma adiponectin concentrations, which may be partly explained by increased adiponectin release from abdominal and gluteal subcutaneous adipose tissue.
Collapse
Affiliation(s)
- Xuewen Wang
- 1Department of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC; 2Department of Exercise and Health Sciences, College of Nursing and Health Sciences, University of Massachusetts Boston, Boston, MA; 3Section on Gerontology and Geriatric Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | | | | | | | | |
Collapse
|
142
|
Wang S, Dougherty EJ, Danner RL. PPARγ signaling and emerging opportunities for improved therapeutics. Pharmacol Res 2016; 111:76-85. [PMID: 27268145 DOI: 10.1016/j.phrs.2016.02.028] [Citation(s) in RCA: 164] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 02/29/2016] [Indexed: 01/23/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated nuclear receptor that regulates glucose and lipid metabolism, endothelial function and inflammation. Rosiglitazone (RGZ) and other thiazolidinedione (TZD) synthetic ligands of PPARγ are insulin sensitizers that have been used for the treatment of type 2 diabetes. However, undesirable side effects including weight gain, fluid retention, bone loss, congestive heart failure, and a possible increased risk of myocardial infarction and bladder cancer, have limited the use of TZDs. Therefore, there is a need to better understand PPARγ signaling and to develop safer and more effective PPARγ-directed therapeutics. In addition to PPARγ itself, many PPARγ ligands including TZDs bind to and activate G protein-coupled receptor 40 (GPR40), also known as free fatty acid receptor 1. GPR40 signaling activates stress kinase pathways that ultimately regulate downstream PPARγ responses. Recent studies in human endothelial cells have demonstrated that RGZ activation of GPR40 is essential to the optimal propagation of PPARγ genomic signaling. RGZ/GPR40/p38 MAPK signaling induces and activates PPARγ co-activator-1α, and recruits E1A binding protein p300 to the promoters of target genes, markedly enhancing PPARγ-dependent transcription. Therefore in endothelium, GPR40 and PPARγ function as an integrated signaling pathway. However, GPR40 can also activate ERK1/2, a proinflammatory kinase that directly phosphorylates and inactivates PPARγ. Thus the role of GPR40 in PPARγ signaling may have important implications for drug development. Ligands that strongly activate PPARγ, but do not bind to or activate GPR40 may be safer than currently approved PPARγ agonists. Alternatively, biased GPR40 agonists might be sought that activate both p38 MAPK and PPARγ, but not ERK1/2, avoiding its harmful effects on PPARγ signaling, insulin resistance and inflammation. Such next generation drugs might be useful in treating not only type 2 diabetes, but also diverse chronic and acute forms of vascular inflammation such as atherosclerosis and septic shock.
Collapse
Affiliation(s)
- Shuibang Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Edward J Dougherty
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robert L Danner
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
143
|
Kautzky-Willer A, Harreiter J, Pacini G. Sex and Gender Differences in Risk, Pathophysiology and Complications of Type 2 Diabetes Mellitus. Endocr Rev 2016; 37:278-316. [PMID: 27159875 PMCID: PMC4890267 DOI: 10.1210/er.2015-1137] [Citation(s) in RCA: 1136] [Impact Index Per Article: 126.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The steep rise of type 2 diabetes mellitus (T2DM) and associated complications go along with mounting evidence of clinically important sex and gender differences. T2DM is more frequently diagnosed at lower age and body mass index in men; however, the most prominent risk factor, which is obesity, is more common in women. Generally, large sex-ratio differences across countries are observed. Diversities in biology, culture, lifestyle, environment, and socioeconomic status impact differences between males and females in predisposition, development, and clinical presentation. Genetic effects and epigenetic mechanisms, nutritional factors and sedentary lifestyle affect risk and complications differently in both sexes. Furthermore, sex hormones have a great impact on energy metabolism, body composition, vascular function, and inflammatory responses. Thus, endocrine imbalances relate to unfavorable cardiometabolic traits, observable in women with androgen excess or men with hypogonadism. Both biological and psychosocial factors are responsible for sex and gender differences in diabetes risk and outcome. Overall, psychosocial stress appears to have greater impact on women rather than on men. In addition, women have greater increases of cardiovascular risk, myocardial infarction, and stroke mortality than men, compared with nondiabetic subjects. However, when dialysis therapy is initiated, mortality is comparable in both males and females. Diabetes appears to attenuate the protective effect of the female sex in the development of cardiac diseases and nephropathy. Endocrine and behavioral factors are involved in gender inequalities and affect the outcome. More research regarding sex-dimorphic pathophysiological mechanisms of T2DM and its complications could contribute to more personalized diabetes care in the future and would thus promote more awareness in terms of sex- and gender-specific risk factors.
Collapse
Affiliation(s)
- Alexandra Kautzky-Willer
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| | - Jürgen Harreiter
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| | - Giovanni Pacini
- Gender Medicine Unit (A.K.-W., J.H.), Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, 1090 Vienna, Austria; and Metabolic Unit (G.P.), Institute of Neuroscience, National Research Council, 35127 Padua, Italy
| |
Collapse
|
144
|
Mesenchymal Stem Cells and Metabolic Syndrome: Current Understanding and Potential Clinical Implications. Stem Cells Int 2016; 2016:2892840. [PMID: 27313625 PMCID: PMC4903149 DOI: 10.1155/2016/2892840] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 04/06/2016] [Accepted: 05/04/2016] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is an obesity-based, complicated clinical condition that has become a global epidemic problem with a high associated risk for cardiovascular disease and mortality. Dyslipidemia, hypertension, and diabetes or glucose dysmetabolism are the major factors constituting metabolic syndrome, and these factors are interrelated and share underlying pathophysiological mechanisms. Severe obesity predisposes individuals to metabolic syndrome, and recent data suggest that mesenchymal stem cells (MSCs) contribute significantly to adipocyte generation by increasing the number of adipocytes. Accordingly, an increasing number of studies have examined the potential roles of MSCs in managing obesity and metabolic syndrome. However, despite the growing bank of experimental and clinical data, the efficacy and the safety of MSCs in the clinical setting are still to be optimized. It is thus hoped that ongoing and future studies can elucidate the roles of MSCs in metabolic syndrome and lead to MSC-based therapeutic options for affected patients. This review discusses current understanding of the relationship between MSCs and metabolic syndrome and its potential implications for patient management.
Collapse
|
145
|
His M, Fagherazzi G, Mesrine S, Boutron-Ruault MC, Clavel-Chapelon F, Dossus L. Prediagnostic body size and breast cancer survival in the E3N cohort study. Int J Cancer 2016; 139:1053-64. [DOI: 10.1002/ijc.30158] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 04/15/2016] [Indexed: 12/18/2022]
Affiliation(s)
- Mathilde His
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| | - Guy Fagherazzi
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| | - Sylvie Mesrine
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| | - Marie-Christine Boutron-Ruault
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| | - Françoise Clavel-Chapelon
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| | - Laure Dossus
- Team 9: Lifestyle, Genes and Health: Integrative Trans-generational Epidemiology, Center for Research in Epidemiology and Population Health; Institut National de la Santé et de la Recherche Médicale (INSERM) U1018; Villejuif France
- University of Paris-Sud; Villejuif France
- Gustave Roussy Institute; Villejuif France
| |
Collapse
|
146
|
Mondal D, Mathur A, Chandra PK. Tripping on TRIB3 at the junction of health, metabolic dysfunction and cancer. Biochimie 2016; 124:34-52. [DOI: 10.1016/j.biochi.2016.02.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 02/04/2016] [Indexed: 12/16/2022]
|
147
|
Mahana D, Trent CM, Kurtz ZD, Bokulich NA, Battaglia T, Chung J, Müller CL, Li H, Bonneau RA, Blaser MJ. Antibiotic perturbation of the murine gut microbiome enhances the adiposity, insulin resistance, and liver disease associated with high-fat diet. Genome Med 2016; 8:48. [PMID: 27124954 PMCID: PMC4847194 DOI: 10.1186/s13073-016-0297-9] [Citation(s) in RCA: 131] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Obesity, type 2 diabetes, and non-alcoholic fatty liver disease (NAFLD) are serious health concerns, especially in Western populations. Antibiotic exposure and high-fat diet (HFD) are important and modifiable factors that may contribute to these diseases. Methods To investigate the relationship of antibiotic exposure with microbiome perturbations in a murine model of growth promotion, C57BL/6 mice received lifelong sub-therapeutic antibiotic treatment (STAT), or not (control), and were fed HFD starting at 13 weeks. To characterize microbiota changes caused by STAT, the V4 region of the 16S rRNA gene was examined from collected fecal samples and analyzed. Results In this model, which included HFD, STAT mice developed increased weight and fat mass compared to controls. Although results in males and females were not identical, insulin resistance and NAFLD were more severe in the STAT mice. Fecal microbiota from STAT mice were distinct from controls. Compared with controls, STAT exposure led to early conserved diet-independent microbiota changes indicative of an immature microbial community. Key taxa were identified as STAT-specific and several were found to be predictive of disease. Inferred network models showed topological shifts concurrent with growth promotion and suggest the presence of keystone species. Conclusions These studies form the basis for new models of type 2 diabetes and NAFLD that involve microbiome perturbation. Electronic supplementary material The online version of this article (doi:10.1186/s13073-016-0297-9) contains supplementary material, which is available to authorized users.
Collapse
|
148
|
Henson J, Edwardson CL, Morgan B, Horsfield MA, Bodicoat DH, Biddle SJH, Gorely T, Nimmo MA, McCann GP, Khunti K, Davies MJ, Yates T. Associations of Sedentary Time with Fat Distribution in a High-Risk Population. Med Sci Sports Exerc 2016; 47:1727-34. [PMID: 25386715 DOI: 10.1249/mss.0000000000000572] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE The effect of sedentary behavior on regional fat deposition, independent of physical activity, remains equivocal. We examined the cross-sectional associations between objectively measured sedentary time and markers of regional fat distribution (heart, liver, visceral, subcutaneous, and total body fat) in a population at a high risk of type 2 diabetes mellitus (T2DM). METHODS Participants were recruited from primary care to two diabetes prevention programs. Sedentary time (<25 counts per 15 s) was measured using ActiGraph GT3X accelerometers. Heart, liver, visceral, subcutaneous, and total body fat were quantified using magnetic resonance images. Fat volumes were calculated by multiplying the cross-sectional areas of the fat-containing pixels by the slice thickness. The liver fat percentage was measured using a representative region of interest created in the right lobe of the liver, avoiding the main portal veins. Linear regression models examined the association of sedentary time with markers of regional fat deposition. RESULTS Sixty-six participants (age, 47.9 ± 16.2 yr; male, 50.0%) were included. After adjustment for several covariates, including glycemia, whole-body fat, and moderate-to-vigorous physical activity, each 30 min of sedentary time was associated with 15.7 cm higher heart fat (P = 0.008), 1.2% higher liver fat (P = 0.026), and 183.7 cm higher visceral fat (P = 0.039). CONCLUSIONS This study provides new evidence suggesting that objectively measured sedentary behavior may have an independent association with heart, liver, and visceral fat in individuals at a high risk of T2DM.
Collapse
Affiliation(s)
- Joseph Henson
- 1National Institute for Health Research Leicester-Loughborough Diet, Lifestyle, and Physical Activity Biomedical Research Unit, and Diabetes Research Centre, College of Medicine, Biological Sciences and Psychology, Leicester, UNITED KINGDOM; 2Department of Cancer Studies and Molecular Medicine, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UNITED KINGDOM; 3Department of Cardiovascular Sciences, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UNITED KINGDOM; 4Institute of Sport, Exercise & Active Living, Victoria University, Melbourne, AUSTRALIA; 5School of Sport, University of Stirling, Stirling, UNITED KINGDOM; 6College of Life and Environmental Sciences, University of Birmingham, Birmingham, UNITED KINGDOM; 7Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Cardiovascular Biomedical Research Unit, Leicester, UNITED KINGDOM; 8National Institute for Health Research Collaborations for Leadership in Applied Health Research and Care East Midlands, and Diabetes Research Centre, College of Medicine, Biological Sciences and Psychology, University of Leicester, Leicester, UNITED KINGDOM
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
149
|
Lin WT, Chan TF, Huang HL, Lee CY, Tsai S, Wu PW, Yang YC, Wang TN, Lee CH. Fructose-Rich Beverage Intake and Central Adiposity, Uric Acid, and Pediatric Insulin Resistance. J Pediatr 2016; 171:90-6.e1. [PMID: 26817591 DOI: 10.1016/j.jpeds.2015.12.061] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 12/02/2015] [Accepted: 12/22/2015] [Indexed: 12/23/2022]
Abstract
OBJECTIVE To determine the association between sugar-sweetened beverage (SSB) consumption with biomarkers of insulin resistance (IR) and investigate whether/how this relates to obesity and serum uric acid in adolescents. STUDY DESIGN Adolescents (n = 1454, aged 12-16 years) were assessed in a study conducted to monitor Multilevel Risk Profiles for Adolescent Metabolic Syndrome in Taiwan. Detailed information about demographics, diet, physical, anthropometric, and clinical variables was collected. An original homeostatic model assessment of IR (HOMA1-IR), updated nonlinear homeostatic model assessment of IR (HOMA2-IR) model, and several IR markers were measured. RESULTS Adolescents who consumed a greater amount of SSBs were more likely to have elevated fasting serum insulin, HOMA1-IR, and HOMA2-IR (P for trends, ≤.028). Compared with SSB nondrinkers, those with >350 mL/d intake of heavy high-fructose corn syrup-containing SSBs had a 0.52 and 0.30 higher multivariate-adjusted HOMA1-IR and HOMA2-IR, respectively. Waist circumference and serum uric acid were correspondingly found to explain 25.4% and 23.6%, as well as 23.2% and 20.6%, of the increases in the 2 IR markers. Both the elevations of HOMA1-IR and HOMA2-IR for high-fructose corn syrup-rich SSB intake were strengthened among obese adolescents (P for interaction, ≤.033). CONCLUSIONS Fructose-rich SSB intake is associated with elevated levels of IR, and this relationship may be partially mediated by central adiposity and serum uric acid. Obesity may modify the effect of this type of SSB consumption in intensifying the elevation of IR in adolescents.
Collapse
Affiliation(s)
- Wei-Ting Lin
- Institute of Environmental and Occupational Health Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Te-Fu Chan
- Department of Obstetrics and Gynecology, Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Hsiao-Ling Huang
- Department of Oral Hygiene, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Ying Lee
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Family Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Sharon Tsai
- Department of Laboratory Medicine, Kaohsiung Municipal Hsiao-Kang Hospital, Kaohsiung, Taiwan
| | - Pei-Wen Wu
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yu-Cheng Yang
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsu-Nai Wang
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Hung Lee
- Department of Public Health, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan; Research Center for Environmental Medicine, Kaohsiung Medical University, Taipei, Taiwan.
| |
Collapse
|
150
|
Olarescu NC, Bollerslev J. The Impact of Adipose Tissue on Insulin Resistance in Acromegaly. Trends Endocrinol Metab 2016; 27:226-237. [PMID: 26948712 DOI: 10.1016/j.tem.2016.02.005] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/11/2023]
Abstract
Adipose tissue (AT) is recognized as key contributor to the systemic insulin resistance and overt diabetes seen in metabolic syndrome. Acromegaly is a disease characterized by excessive secretion of growth hormone (GH) and insulin-like growth factor I (IGF-I). GH is known both for its action on AT and for its detrimental effect on glucose metabolism and insulin signaling. In active acromegaly, while body fat deports are diminished, insulin resistance is increased. Early studies have demonstrated defects in insulin action, both at the hepatic and extrahepatic (i.e., muscle and fat) levels, in active disease. This review discusses recent data suggesting that AT inflammation, altered AT distribution, and impaired adipogenesis are potential mechanisms contributing to systemic insulin resistance in acromegaly.
Collapse
Affiliation(s)
- Nicoleta Cristina Olarescu
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway.
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Department of Endocrinology, Oslo University Hospital, Rikshospitalet, Oslo, Norway; Faculty of Medicine, University of Oslo, Norway
| |
Collapse
|