301
|
Moysidou M, Karaliota S, Kodela E, Salagianni M, Koutmani Y, Katsouda A, Kodella K, Tsakanikas P, Ourailidou S, Andreakos E, Kostomitsopoulos N, Skokos D, Chatzigeorgiou A, Chung KJ, Bornstein S, Sleeman MW, Chavakis T, Karalis KP. CD8+ T cells in beige adipogenesis and energy homeostasis. JCI Insight 2018; 3:e95456. [PMID: 29515042 PMCID: PMC5922290 DOI: 10.1172/jci.insight.95456] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 02/01/2018] [Indexed: 12/19/2022] Open
Abstract
Although accumulation of lymphocytes in the white adipose tissue (WAT) in obesity is linked to insulin resistance, it remains unclear whether lymphocytes also participate in the regulation of energy homeostasis in the WAT. Here, we demonstrate enhanced energy dissipation in Rag1-/- mice, increased catecholaminergic input to subcutaneous WAT, and significant beige adipogenesis. Adoptive transfer experiments demonstrated that CD8+ T cell deficiency accounts for the enhanced beige adipogenesis in Rag1-/- mice. Consistently, we identified that CD8-/- mice also presented with enhanced beige adipogenesis. The inhibitory effect of CD8+ T cells on beige adipogenesis was reversed by blockade of IFN-γ. All together, our findings identify an effect of CD8+ T cells in regulating energy dissipation in lean WAT, mediated by IFN-γ modulation of the abundance of resident immune cells and of local catecholaminergic activity. Our results provide a plausible explanation for the clinical signs of metabolic dysfunction in diseases characterized by altered CD8+ T cell abundance and suggest targeting of CD8+ T cells as a promising therapeutic approach for obesity and other diseases with altered energy homeostasis.
Collapse
MESH Headings
- Adipogenesis/physiology
- Adipose Tissue, Beige/cytology
- Adipose Tissue, Beige/immunology
- Adipose Tissue, Beige/metabolism
- Adipose Tissue, White/cytology
- Adipose Tissue, White/immunology
- Adipose Tissue, White/metabolism
- Adoptive Transfer
- Animals
- CD8 Antigens/genetics
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Differentiation/physiology
- Disease Models, Animal
- Energy Metabolism/immunology
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Interferon-gamma/genetics
- Interferon-gamma/metabolism
- Lipid Metabolism/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Models, Animal
- Obesity/genetics
- Obesity/immunology
- Obesity/metabolism
Collapse
Affiliation(s)
- Maria Moysidou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- University of Crete, School of Medicine, Heraklion, Crete, Greece
| | - Sevasti Karaliota
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Elisavet Kodela
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- University of Crete, School of Medicine, Heraklion, Crete, Greece
| | - Maria Salagianni
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Yassemi Koutmani
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Antonia Katsouda
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Konstantia Kodella
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Panagiotis Tsakanikas
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Styliani Ourailidou
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Evangelos Andreakos
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Nikolaos Kostomitsopoulos
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | | | | | - Kyoung-Jin Chung
- Technische Universität Dresden, School of Medicine, Dresden, Germany
| | - Stefan Bornstein
- Technische Universität Dresden, School of Medicine, Dresden, Germany
| | - Mark W. Sleeman
- Department of Physiology, Monash University, Clayton, Victoria, Australia
| | | | - Katia P. Karalis
- Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation Academy of Athens, Athens, Greece
- Technische Universität Dresden, School of Medicine, Dresden, Germany
- Endocrine Division, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
302
|
Jastroch M, Oelkrug R, Keipert S. Insights into brown adipose tissue evolution and function from non-model organisms. ACTA ACUST UNITED AC 2018. [PMID: 29514888 DOI: 10.1242/jeb.169425] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Brown adipose tissue (BAT) enables adaptive thermoregulation through heat production that is catalyzed by mitochondrial uncoupling protein 1 (UCP1). BAT is frequently studied in rodent model organisms, and recently in adult humans to treat metabolic diseases. However, complementary studies of many non-model species, which have diversified to many more ecological niches, may significantly broaden our understanding of BAT regulation and its physiological roles. This Review highlights the research on non-model organisms, which was instrumental to the discovery of BAT function, and the unique evolutionary history of BAT/UCP1 in mammalian thermogenesis. The comparative biology of BAT provides a powerful integrative approach that could identify conserved and specialized functional changes in BAT and UCP1 by considering species diversity, ecology and evolution, and by fusing multiple scientific disciplines such as physiology and biochemistry. Thus, resolving the complete picture of BAT biology may fail if comparative studies of non-model organisms are neglected.
Collapse
Affiliation(s)
- Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, D-85764 Neuherberg, Germany .,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany.,Department of Animal Physiology, Faculty of Biology, Philipps University of Marburg, D-35032 Marburg, Germany
| | - Rebecca Oelkrug
- Department of Molecular Endocrinology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, D-23562 Lübeck, Germany
| | - Susanne Keipert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, D-85764 Neuherberg, Germany.,German Center for Diabetes Research (DZD), D-85764 Neuherberg, Germany
| |
Collapse
|
303
|
Abstract
The beneficial effects of exercise on skeletal muscle and the cardiovascular system have long been known. Recent studies have focused on investigating the effects of exercise on adipose tissue and the effects that these exercise-induced adaptations have on overall metabolic health. Examination of exercise-induced adaptations in both white adipose tissue (WAT) and brown adipose tissue (BAT) has revealed marked differences in each tissue with exercise. In WAT, there are changes to both subcutaneous WAT (scWAT) and visceral WAT (vWAT), including decreased adipocyte size and lipid content, increased expression of metabolic genes, altered secretion of adipokines and increased mitochondrial activity. Adaptations specific to scWAT include lipidomic remodeling of phospholipids and, in rodents, the beiging of scWAT. The changes to BAT are less clear: studies evaluating the effect of exercise on the BAT of humans and rodents have revealed contradictory data, making this an important area of current investigation. In this Review, we discuss the exercise-induced changes to WAT and BAT that have been reported by different studies and highlight the current questions in this field.
Collapse
Affiliation(s)
- Adam C Lehnig
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210, USA
| | - Kristin I Stanford
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio 43210, USA
| |
Collapse
|
304
|
Abstract
Brown adipose tissue (BAT) thermogenesis is critical for thermoregulation and contributes to total energy expenditure. However, whether BAT has non-thermogenic functions is largely unknown. Here, we describe that BAT-specific liver kinase b1 knockout (Lkb1BKO) mice exhibited impaired BAT mitochondrial respiration and thermogenesis but reduced adiposity and liver triglyceride accumulation under high-fat-diet feeding at room temperature. Importantly, these metabolic benefits were also present in Lkb1BKO mice at thermoneutrality, where BAT thermogenesis was not required. Mechanistically, decreased mRNA levels of mtDNA-encoded electron transport chain (ETC) subunits and ETC proteome imbalance led to defective BAT mitochondrial respiration in Lkb1BKO mice. Furthermore, reducing mtDNA gene expression directly in BAT by removing mitochondrial transcription factor A (Tfam) in BAT also showed ETC proteome imbalance and the trade-off between BAT thermogenesis and systemic metabolism at room temperature and thermoneutrality. Collectively, our data demonstrate that ETC proteome imbalance in BAT regulates systemic metabolism independently of thermogenesis.
Collapse
|
305
|
Lim S, Hosaka K, Nakamura M, Cao Y. Co-option of pre-existing vascular beds in adipose tissue controls tumor growth rates and angiogenesis. Oncotarget 2018; 7:38282-38291. [PMID: 27203675 PMCID: PMC5122389 DOI: 10.18632/oncotarget.9436] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 04/27/2016] [Indexed: 12/15/2022] Open
Abstract
Many types of cancer develop in close association with highly vascularized adipose tissues. However, the role of adipose pre-existing vascular beds on tumor growth and angiogenesis is unknown. Here we report that pre-existing microvascular density in tissues where tumors originate is a crucial determinant for tumor growth and neovascularization. In three independent tumor types including breast cancer, melanoma, and fibrosarcoma, inoculation of tumor cells in the subcutaneous tissue, white adipose tissue (WAT), and brown adipose tissue (BAT) resulted in markedly differential tumor growth rates and angiogenesis, which were in concordance with the degree of pre-existing vascularization in these tissues. Relative to subcutaneous tumors, WAT and BAT tumors grew at accelerated rates along with improved neovascularization, blood perfusion, and decreased hypoxia. Tumor cells implanted in adipose tissues contained leaky microvessel with poor perivascular cell coverage. Thus, adipose vasculature predetermines the tumor microenvironment that eventually supports tumor growth.
Collapse
Affiliation(s)
- Sharon Lim
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Kayoko Hosaka
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Masaki Nakamura
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, 171 77 Stockholm, Sweden.,Department of Medical and Health Sciences, Linköping University, 581 83 Linköping, Sweden.,Affiliated WuXi No 2 Hospital of Nanjing Medical University, 214 002 Wuxi, China.,Department of Cardiovascular Sciences, University of Leicester and NIHR Leicester Cardiovascular Biomedical Research Unit, Glenfield Hospital, LE3 9QP Leicester, UK
| |
Collapse
|
306
|
Sepa-Kishi DM, Ceddia RB. White and beige adipocytes: are they metabolically distinct? Horm Mol Biol Clin Investig 2018; 33:/j/hmbci.ahead-of-print/hmbci-2018-0003/hmbci-2018-0003.xml. [PMID: 29466235 DOI: 10.1515/hmbci-2018-0003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/22/2018] [Indexed: 12/17/2022]
Abstract
The white adipose tissue (WAT) exhibits great plasticity and can undergo "browning" and acquire features of the brown adipose tissue (BAT), which takes place following cold exposure, chronic endurance exercise or β3-adrenergic stimulation. WAT that underwent browning is characterized by the presence of "beige" adipocytes, which are morphologically similar to brown adipocytes, express uncoupling protein 1 (UCP1) and are considered thermogenically competent. Thus, inducing a BAT-like phenotype in the WAT could promote energy dissipation within this depot, reducing the availability of substrate that would otherwise be stored in the WAT. Importantly, BAT in humans only represents a small proportion of total body mass, which limits the thermogenic capacity of this tissue. Therefore, browning of the WAT could significantly expand the energy-dissipating capacity of the organism and be of therapeutic value in the treatment of metabolic diseases. However, the question remains as to whether WAT indeed changes its metabolic profile from an essentially fat storage/release compartment to an energy dissipating compartment that functions much like BAT. Here, we discuss the differences with respect to thermogenic capacity and metabolic characteristics between white and beige adipocytes to determine whether the latter cells indeed significantly enhance their capacity to dissipate energy through UCP1-mediated mitochondrial uncoupling or by the activation of alternative UCP1-independent futile cycles.
Collapse
Affiliation(s)
- Diane M Sepa-Kishi
- Muscle Health Research Center, School of Kinesiology and Health Science, York University, Toronto, Canada
| | - Rolando B Ceddia
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, 4700 Keele St., North York, Ontario, M3J 13P, Canada, Phone: 416-736-2100 (Ext. 77204), Fax: 416-736-5774
| |
Collapse
|
307
|
Chechi K, van Marken Lichtenbelt W, Richard D. Brown and beige adipose tissues: phenotype and metabolic potential in mice and men. J Appl Physiol (1985) 2018; 124:482-496. [PMID: 28302705 PMCID: PMC5867364 DOI: 10.1152/japplphysiol.00021.2017] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/09/2017] [Accepted: 03/13/2017] [Indexed: 01/06/2023] Open
Abstract
With the recent rediscovery of brown fat in adult humans, our outlook on adipose tissue biology has undergone a paradigm shift. While we attempt to identify, recruit, and activate classic brown fat stores in humans, identification of beige fat has also raised the possibility of browning our white fat stores. Whether such transformation of human white fat depots can be achieved to enhance the whole body oxidative potential remains to be seen. Evidence to date, however, largely points toward a major oxidative role only for classic brown fat depots, at least in rodents. White fat stores seem to provide the main fuel for sustaining thermogenesis via lipolysis. Interestingly, molecular markers consistent with both classic brown and beige fat identity can be observed in human supraclavicular depot, thereby complicating the discussion on beige fat in humans. Here, we review the recent advances made in our understanding of brown and beige fat in humans and mice. We further provide an overview of their plausible physiological relevance to whole body energy metabolism.
Collapse
Affiliation(s)
- Kanta Chechi
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| | - Wouter van Marken Lichtenbelt
- Department of Human Biology, NUTRIM School for Nutrition, Toxicology and Metabolism, Maastricht University Medical Center , Maastricht , The Netherlands
| | - Denis Richard
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Ville de Québec, Quebec , Canada
| |
Collapse
|
308
|
Abstract
Brown fat is emerging as an interesting and promising target for therapeutic intervention in obesity and metabolic disease. Activation of brown fat in humans is associated with marked improvement in metabolic parameters such as levels of free fatty acids and insulin sensitivity. Skeletal muscle is another important organ for thermogenesis, with the capacity to induce energy-consuming futile cycles. In this Review, we focus on how these two major thermogenic organs - brown fat and muscle - act and cooperate to maintain normal body temperature. Moreover, in the light of disease-relevant mechanisms, we explore the molecular pathways that regulate thermogenesis in brown fat and muscle. Brown adipocytes possess a unique cellular mechanism to convert chemical energy into heat: uncoupling protein 1 (UCP1), which can short-circuit the mitochondrial proton gradient. However, recent research demonstrates the existence of several other energy-expending 'futile' cycles in both adipocytes and muscle, such as creatine and calcium cycling. These mechanisms can complement or even substitute for UCP1-mediated thermogenesis. Moreover, they expand our view of cold-induced thermogenesis from a special feature of brown adipocytes to a more general physiological principle. Finally, we discuss how thermogenic mechanisms can be exploited to expend energy and hence offer new therapeutic opportunities.
Collapse
Affiliation(s)
- Matthias J Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Sven Enerbäck
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, The Sahlgrenska Academy, University of Gothenburg, Medicinaregatan 9A, SE-405 30 Gothenburg, Sweden
| |
Collapse
|
309
|
Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, Zhang Q, Guo C, Zhang L, Wang Q. Exosomes From Adipose-Derived Stem Cells Attenuate Adipose Inflammation and Obesity Through Polarizing M2 Macrophages and Beiging in White Adipose Tissue. Diabetes 2018; 67:235-247. [PMID: 29133512 DOI: 10.2337/db17-0356] [Citation(s) in RCA: 472] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 11/04/2017] [Indexed: 12/15/2022]
Abstract
Adipose-derived stem cells (ADSCs) play critical roles in controlling obesity-associated inflammation and metabolic disorders. Exosomes from ADSCs exert protective effects in several diseases, but their roles in obesity and related pathological conditions remain unclear. In this study, we showed that treatment of obese mice with ADSC-derived exosomes facilitated their metabolic homeostasis, including improved insulin sensitivity (27.8% improvement), reduced obesity, and alleviated hepatic steatosis. ADSC-derived exosomes drove alternatively activated M2 macrophage polarization, inflammation reduction, and beiging in white adipose tissue (WAT) of diet-induced obese mice. Mechanistically, exosomes from ADSCs transferred into macrophages to induce anti-inflammatory M2 phenotypes through the transactivation of arginase-1 by exosome-carried active STAT3. Moreover, M2 macrophages induced by ADSC-derived exosomes not only expressed high levels of tyrosine hydroxylase responsible for catecholamine release, but also promoted ADSC proliferation and lactate production, thereby favoring WAT beiging and homeostasis in response to high-fat challenge. These findings delineate a novel exosome-mediated mechanism for ADSC-macrophage cross talk that facilitates immune and metabolic homeostasis in WAT, thus providing potential therapy for obesity and diabetes.
Collapse
MESH Headings
- Adipocytes, Beige/immunology
- Adipocytes, Beige/metabolism
- Adipocytes, Beige/pathology
- Adipocytes, White/immunology
- Adipocytes, White/metabolism
- Adipocytes, White/pathology
- Adipogenesis
- Adult Stem Cells/immunology
- Adult Stem Cells/metabolism
- Adult Stem Cells/pathology
- Animals
- Biomarkers/metabolism
- Cell Communication
- Cell Polarity
- Cell Proliferation
- Cells, Cultured
- Diet, High-Fat/adverse effects
- Exosomes/immunology
- Exosomes/metabolism
- Exosomes/pathology
- Exosomes/transplantation
- Insulin Resistance
- Macrophage Activation
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Macrophages, Peritoneal/pathology
- Macrophages, Peritoneal/transplantation
- Male
- Mice, Inbred C57BL
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Non-alcoholic Fatty Liver Disease/etiology
- Non-alcoholic Fatty Liver Disease/prevention & control
- Obesity/immunology
- Obesity/pathology
- Obesity/physiopathology
- Obesity/therapy
- Phagocytosis
Collapse
Affiliation(s)
- Hui Zhao
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Qianwen Shang
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Zhenzhen Pan
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Yang Bai
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Zequn Li
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Huiying Zhang
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Qiu Zhang
- School of Environmental Science and Engineering, Shandong University, Jinan, Shandong, China
| | - Chun Guo
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Lining Zhang
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Qun Wang
- Department of Immunology and Key Laboratory of Infection and Immunity of Shandong Province, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| |
Collapse
|
310
|
Zaiou M, El Amri H, Bakillah A. The clinical potential of adipogenesis and obesity-related microRNAs. Nutr Metab Cardiovasc Dis 2018; 28:91-111. [PMID: 29170059 DOI: 10.1016/j.numecd.2017.10.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/12/2017] [Accepted: 10/15/2017] [Indexed: 02/07/2023]
Abstract
Obesity is a growing health problem commonly associated with numerous metabolic disorders including type 2 diabetes, hypertension, cardiovascular disease, and some forms of cancer. The burden of obesity and associated cardiometabolic diseases are believed to arise through complex interplay between genetics and epigenetics predisposition, nutrition, environment, and lifestyle. However, the molecular basis and the repertoire of obesity-affecting factors are still unknown. Emerging evidence is connecting microRNAs (miRNAs) dysregulation with adipogenesis and obesity. Alteration in miRNAs expression could result in changes in the pattern of genes controlling a range of biological processes including inflammation, lipid metabolism, insulin resistance and adipogenesis. Hence, understanding exact roles of miRNAs as well as the degree of their contribution to the regulation of adipogenesis and fat cell development in obesity would provide new therapeutic targets for the development of novel and effective anti-obesity drugs. The objective of the current review is to: (i) discuss some of the latest development on relevant miRNAs dysregulation mainly in human adipogenesis and obesity, (ii) emphasize the role of circulating miRNAs as new promising therapeutics and attractive potential biomarkers for treating obesity and associated risk factor diseases, (iii) describe how dietary factors may influence obesity through modulation of miRNAs expression, (iv) highlight some of the actual limitations to the promise of miRNAs as novel therapeutics as well as to their translation for the benefit of patients, and finally (v) provide recommendations for future research on miRNA-based therapeutics that could lead to a breakthrough in the treatment of obesity and its associated pathologies.
Collapse
Affiliation(s)
- M Zaiou
- Université de Lorraine, Faculté de Pharmacie, 5 rue Albert Lebrun, 54000, Nancy, France.
| | - H El Amri
- Laboratoire de Génétique de la Gendarmerie Royale, Avenue Ibn Sina, Agdal, Rabat, Morocco
| | - A Bakillah
- State University of New York, Downstate Medical Center, Department of Medicine, 450 Clarkson Ave., Brooklyn, NY, 11203, USA
| |
Collapse
|
311
|
Law J, Chalmers J, Morris DE, Robinson L, Budge H, Symonds ME. The use of infrared thermography in the measurement and characterization of brown adipose tissue activation. Temperature (Austin) 2018; 5:147-161. [PMID: 30393752 DOI: 10.1080/23328940.2017.1397085] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 10/16/2017] [Accepted: 10/23/2017] [Indexed: 10/18/2022] Open
Abstract
Interest in brown adipose tissue has increased in recent years as a potential target for novel obesity, diabetes and metabolic disease treatments. One of the significant limitations to rapid progress has been the difficulty in measuring brown adipose tissue activity, especially in humans. Infrared thermography (IRT) is being increasingly recognized as a valid and complementary method to standard imaging modalities, such as positron emission tomography-computed tomography (PET/CT). In contrast to PET/CT, it is non-invasive, cheap and quick, allowing, for the first time, the possibility of large studies of brown adipose tissue (BAT) on healthy populations and children. Variations in study protocols and analysis methods currently limit direct comparison between studies but IRT following appropriate BAT stimulation consistently shows a change in supraclavicular skin temperature and a close association with results from BAT measurements from other methods.
Collapse
Affiliation(s)
- James Law
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Jane Chalmers
- Nottingham Digestive Diseases Centre, University of Nottingham and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham
| | - David E Morris
- Department of Electrical & Electronic Engineering, Faculty of Engineering, University of Nottingham, United Kingdom
| | - Lindsay Robinson
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Helen Budge
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Michael E Symonds
- Early Life Research Unit, Division of Child Health, Obstetrics & Gynaecology, School of Medicine, University of Nottingham, Nottingham, United Kingdom.,Nottingham Digestive Diseases Centre, University of Nottingham and National Institute for Health Research (NIHR) Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham
| |
Collapse
|
312
|
Kuda O, Rossmeisl M, Kopecky J. Omega-3 fatty acids and adipose tissue biology. Mol Aspects Med 2018; 64:147-160. [PMID: 29329795 DOI: 10.1016/j.mam.2018.01.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 01/05/2018] [Accepted: 01/08/2018] [Indexed: 12/16/2022]
Abstract
This review provides evidence for the importance of white and brown adipose tissue (i.e. WAT and BAT) function for the maintenance of healthy metabolic phenotype and its preservation in response to omega-3 polyunsaturated fatty acids (omega-3 PUFA), namely in the context of diseased states linked to aberrant accumulation of body fat, systemic low-grade inflammation, dyslipidemia and insulin resistance. More specifically, the review deals with (i) the concept of immunometabolism, i.e. how adipose-resident immune cells and adipocytes affect each other and define the immune-metabolic interface; and (ii) the characteristic features of "healthy adipocytes" in WAT, which are relatively small fat cells endowed with a high capacity for mitochondrial oxidative phosphorylation, triacylglycerol/fatty acid (TAG/FA) cycling and de novo lipogenesis (DNL). The intrinsic metabolic features of WAT and their flexible regulations, reflecting the presence of "healthy adipocytes", provide beneficial local and systemic effects, including (i) protection against in situ endoplasmic reticulum stress and related inflammatory response during activation of adipocyte lipolysis; (ii) prevention of ectopic fat accumulation and dyslipidemia caused by increased hepatic VLDL synthesis, as well as prevention of lipotoxic damage of insulin signaling in extra-adipose tissues; and also (iii) increased synthesis of anti-inflammatory and insulin-sensitizing lipid mediators with pro-resolving properties, including the branched fatty acid esters of hydroxy fatty acids (FAHFAs), also depending on the activity of DNL in WAT. The "healthy adipocytes" phenotype can be induced in WAT of obese mice in response to various stimuli including dietary omega-3 PUFA, especially when combined with moderate calorie restriction, and possibly also with other life style (e.g. physical activity) or pharmacological (e.g. thiazolidinediones) interventions. While omega-3 PUFA could exert beneficial systemic effects by improving immunometabolism of WAT without a concomitant induction of BAT, it is currently not clear whether the metabolic effects of the combined intervention using omega-3 PUFA and calorie restriction or thiazolidinediones depend also on the activation of BAT function and/or the induction of brite/beige adipocytes in WAT. It remains to be established why omega-3 PUFA intervention in type 2 diabetic subjects does not improve insulin sensitivity and glucose homeostasis despite inducing various anti-inflammatory mediators in WAT, including the recently discovered docosahexaenoyl esters of hydroxy linoleic acid, the lipokines from the FAHFA family, as well as several endocannabinoid-related anti-inflammatory lipids. To answer the question whether and to which extent omega-3 PUFA supplementation could promote the formation of "healthy adipocytes" in WAT of human subjects, namely in the obese insulin-resistant patients, represents a challenging task that is of great importance for the treatment of some serious non-communicable diseases.
Collapse
Affiliation(s)
- Ondrej Kuda
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Martin Rossmeisl
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic
| | - Jan Kopecky
- Department of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska, 1083 Prague 4, Czech Republic.
| |
Collapse
|
313
|
TGR5 signalling promotes mitochondrial fission and beige remodelling of white adipose tissue. Nat Commun 2018; 9:245. [PMID: 29339725 PMCID: PMC5770450 DOI: 10.1038/s41467-017-02068-0] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 11/03/2017] [Indexed: 01/03/2023] Open
Abstract
Remodelling of energy storing white fat into energy expending beige fat could be a promising strategy to reduce adiposity. Here, we show that the bile acid-responsive membrane receptor TGR5 mediates beiging of the subcutaneous white adipose tissue (scWAT) under multiple environmental cues including cold exposure and prolonged high-fat diet feeding. Moreover, administration of TGR5-selective bile acid mimetics to thermoneutral housed mice leads to the appearance of beige adipocyte markers and increases mitochondrial content in the scWAT of Tgr5+/+ mice but not in their Tgr5−/− littermates. This phenotype is recapitulated in vitro in differentiated adipocytes, in which TGR5 activation increases free fatty acid availability through lipolysis, hence fuelling β-oxidation and thermogenic activity. TGR5 signalling also induces mitochondrial fission through the ERK/DRP1 pathway, further improving mitochondrial respiration. Taken together, these data identify TGR5 as a druggable target to promote beiging with potential applications in the management of metabolic disorders. White adipose tissue can undergo a process of beiging and acquire functional characteristics similar to brown adipose tissue, including the ability to dissipate energy via uncoupled respiration. Here, Velazquez-Villegas et al. show that activation of the bile acid membrane receptor, TGR5, leads to white adipocyte beiging by promoting mitochondrial fission.
Collapse
|
314
|
Abstract
The chaperone GRP78 (glucose related protein 78), also called BiP (binding immunoglobulin protein) is a key regulator of endoplasmic reticulum (ER) stress. We recently described that over-expression of GRP78 specifically in the ventromedial nucleus of the hypothalamus (VMH) releases hypothalamic ER stress in rodent obese models leading to weight loss, reduced hepatic steatosis and improved insulin and leptin sensitivity. The action of GRP78 is mediated by a feeding-independent mechanism involving increased sympathetic tone, augmented brown adipose tissue (BAT) thermogenesis and induction browning of white adipose tissue (WAT).
Collapse
Affiliation(s)
- Cristina Contreras
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela, -Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Marcos F. Fondevila
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela, -Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| | - Miguel López
- NeurObesity Group, Department of Physiology, CIMUS, University of Santiago de Compostela, -Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain
| |
Collapse
|
315
|
Pisani DF, Barquissau V, Chambard JC, Beuzelin D, Ghandour RA, Giroud M, Mairal A, Pagnotta S, Cinti S, Langin D, Amri EZ. Mitochondrial fission is associated with UCP1 activity in human brite/beige adipocytes. Mol Metab 2018; 7:35-44. [PMID: 29198749 PMCID: PMC5784321 DOI: 10.1016/j.molmet.2017.11.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE Thermogenic adipocytes (i.e. brown or brite/beige adipocytes) are able to burn large amounts of lipids and carbohydrates as a result of highly active mitochondria and enhanced uncoupled respiration, due to UCP1 activity. Although mitochondria are the key organelles for this thermogenic function, limited human data are available. METHODS/RESULTS We characterized changes in the mitochondrial function of human brite adipocytes, using hMADS cells as a model of white- to brite-adipocyte conversion. We found that profound molecular modifications were associated with morphological changes in mitochondria. The fission process was partly driven by the DRP1 protein, which also promoted mitochondrial uncoupling. CONCLUSION Our data demonstrate that white-to-brite conversion of human adipocytes relies on molecular, morphological and functional changes in mitochondria, which enable brite/beige cells to carry out thermogenesis.
Collapse
Affiliation(s)
| | - Valentin Barquissau
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse 31432, France
| | | | - Diane Beuzelin
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse 31432, France
| | | | - Maude Giroud
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France
| | - Aline Mairal
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse 31432, France
| | - Sophie Pagnotta
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France; Centre Commun de Microscopie Appliquée, Microscopy and Imaging Platform Côte d'Azur, Nice 06108, France
| | - Saverio Cinti
- Department of Experimental and Clinical Medicine, University of Ancona (Politecnica Delle Marche), Ancona 60121, Italy; Center of Obesity, University of Ancona (Politecnica Delle Marche)-United Hospitals, Ancona 60121, Italy
| | - Dominique Langin
- Inserm, UMR1048, Obesity Research Laboratory, Institute of Metabolic and Cardiovascular Diseases, Toulouse 31432, France; University of Toulouse, UMR1048, Paul Sabatier University, Toulouse 31432, France; Toulouse University Hospitals, Department of Clinical Biochemistry, Toulouse 31059, France
| | - Ez-Zoubir Amri
- Université Côte d'Azur, CNRS, Inserm, iBV, Nice, France.
| |
Collapse
|
316
|
Haenisch M, Treuting PM, Brabb T, Goldstein AS, Berkseth K, Amory JK, Paik J. Pharmacological inhibition of ALDH1A enzymes suppresses weight gain in a mouse model of diet-induced obesity. Obes Res Clin Pract 2018; 12:93-101. [PMID: 28919001 PMCID: PMC5816716 DOI: 10.1016/j.orcp.2017.08.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 08/19/2017] [Accepted: 08/22/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Retinoic acid (RA) is known to play a role in weight regulation. Because mice without ALDH1A1, a major RA synthesizing enzyme, are resistant to diet-induced obesity, we tested a hypothesis that pharmacological inhibition of RA synthesis can suppress weight gain in a murine model of diet-induced obesity. METHODS C57BL/6J male mice were fed a high fat diet (HFD) for 8 weeks to induce obesity and then randomized to a HFD with or without WIN 18,446, an RA synthesis inhibitor, for an additional 9 weeks. Body weight, body composition, energy expenditure, activity, and food intake were measured. Levels of retinoids, lipids, and genes involved in the metabolism of retinoid and lipids were also determined. RESULT s Mice treated with WIN 18,446 gained significantly less weight and had decreased adipose tissue weight, adipocyte size, and macrophage infiltration in adipose tissue. In addition, we observed higher UCP1 expression in adipose tissues and decreased expression of RA responsive genes and genes involved in fatty acid synthesis in the livers and lungs of mice treated with WIN 18,446. CONCLUSIONS Pharmacological suppression of RA synthesis via inhibition of ALDH1A1 may be a potential target for treatment of obesity.
Collapse
Affiliation(s)
- Michael Haenisch
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Piper M Treuting
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | - Thea Brabb
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA
| | | | - Kathryn Berkseth
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - John K Amory
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jisun Paik
- Department of Comparative Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|
317
|
Merlin J, Sato M, Chia LY, Fahey R, Pakzad M, Nowell CJ, Summers RJ, Bengtsson T, Evans BA, Hutchinson DS. Rosiglitazone and a β 3-Adrenoceptor Agonist Are Both Required for Functional Browning of White Adipocytes in Culture. Front Endocrinol (Lausanne) 2018; 9:249. [PMID: 29910772 PMCID: PMC5992408 DOI: 10.3389/fendo.2018.00249] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023] Open
Abstract
The recruitment of brite (or beige) adipocytes has been advocated as a means to combat obesity, due to their ability to phenotypically resemble brown adipocytes (BA). Lineage studies indicate that brite adipocytes are formed by differentiation of precursor cells or by direct conversion of existing white adipocytes, depending on the adipose depot examined. We have systematically compared the gene expression profile and a functional output (oxygen consumption) in mouse adipocytes cultured from two contrasting depots, namely interscapular brown adipose tissue, and inguinal white adipose tissue (iWAT), following treatment with a known browning agent, the peroxisome proliferator-activated receptor (PPARγ) activator rosiglitazone. Prototypical BA readily express uncoupling protein (UCP)1, and upstream regulators including the β3-adrenoceptor and transcription factors involved in energy homeostasis. Adipocytes from inguinal WAT display maximal UCP1 expression and mitochondrial uncoupling only when treated with a combination of the PPARγ activator rosiglitazone and a β3-adrenoceptor agonist. In conclusion, brite adipocytes are fully activated only when a browning agent (rosiglitazone) and a thermogenic agent (β3-adrenoceptor agonist) are added in combination. The presence of rosiglitazone throughout the 7-day culture period partially masks the effects of β3-adrenoceptor signaling in inguinal white adipocyte cultures, whereas including rosiglitazone only for the first 3 days promotes robust β3-adrenoceptor expression and provides an improved window for detection of β3-adrenoceptor responses.
Collapse
Affiliation(s)
- Jon Merlin
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Masaaki Sato
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Richard Fahey
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Mohsen Pakzad
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Cameron J. Nowell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Roger J. Summers
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Bronwyn A. Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Dana S. Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
- Department of Pharmacology, Monash University, Clayton, VIC, Australia
- *Correspondence: Dana S. Hutchinson,
| |
Collapse
|
318
|
Cioffi F, Gentile A, Silvestri E, Goglia F, Lombardi A. Effect of Iodothyronines on Thermogenesis: Focus on Brown Adipose Tissue. Front Endocrinol (Lausanne) 2018; 9:254. [PMID: 29875734 PMCID: PMC5974034 DOI: 10.3389/fendo.2018.00254] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 05/03/2018] [Indexed: 02/05/2023] Open
Abstract
Thyroid hormones significantly influence energy expenditure by affecting the activity of metabolic active tissues, among which, mammalian brown adipose tissue (BAT) plays a significant role. For a long time, the modulation of BAT activity by 3,3',5-triiodo-l-thyronine (T3) has been ascribed to its direct actions on this tissue; however, recent evidence indicates that T3, by stimulating specific brain centers, activates the metabolism of BAT via the sympathetic nervous system. These distinct mechanisms of action are not mutually exclusive. New evidence indicates that 3,5-diiodo-l-thyronine (3,5-T2), a thyroid hormone derivative, exerts thermogenic effects, by influencing mitochondrial activity in metabolically active tissues, such as liver, skeletal muscle, and BAT. At the moment, due to the absence of experiments finalized to render a clear cut discrimination between peripheral and central effects induced by 3,5-T2, it is not possible to exclude that some of the metabolic effects exerted by 3,5-T2 may be mediated centrally. Despite this, some evidence suggests that 3,5-T2 plays a role in adrenergic stimulation of thermogenesis in BAT. This mini-review provides an overview of the effects induced by T3 and 3,5-T2 on BAT thermogenesis, with a focus on data suggesting the involvement of central adrenergic stimulation. These aspects may reveal new perspectives in thyroid physiology and in the control of energy metabolism.
Collapse
Affiliation(s)
- Federica Cioffi
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | | | - Elena Silvestri
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
| | - Fernando Goglia
- Department of Sciences and Technologies, University of Sannio, Benevento, Italy
- *Correspondence: Fernando Goglia, ; Assunta Lombardi,
| | - Assunta Lombardi
- Department of Biology, University of Naples Federico II, Naples, Italy
- *Correspondence: Fernando Goglia, ; Assunta Lombardi,
| |
Collapse
|
319
|
Karunakaran S, Clee SM. Genetics of metabolic syndrome: potential clues from wild-derived inbred mouse strains. Physiol Genomics 2018; 50:35-51. [DOI: 10.1152/physiolgenomics.00059.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The metabolic syndrome (MetS) is a complex constellation of metabolic abnormalities including obesity, abnormal glucose metabolism, dyslipidemia, and elevated blood pressure that together substantially increase risk for cardiovascular disease and Type 2 diabetes. Both genetic and environmental factors contribute to the development of MetS, but this process is still far from understood. Human studies have revealed only part of the underlying basis. Studies in mice offer many strengths that can complement human studies to help elucidate the etiology and pathophysiology of MetS. Here we review the ways mice can contribute to MetS research. In particular, we focus on the information that can be obtained from studies of the inbred strains, with specific focus on the phenotypes of the wild-derived inbred strains. These are newly derived inbred strains that were created from wild-caught mice. They contain substantial genetic variation that is not present in the classical inbred strains, have phenotypes of relevance for MetS, and various mouse strain resources have been created to facilitate the mining of this new genetic variation. Thus studies using wild-derived inbred strains hold great promise for increasing our understanding of MetS.
Collapse
Affiliation(s)
- Subashini Karunakaran
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Susanne M. Clee
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
320
|
Fruhwürth S, Vogel H, Schürmann A, Williams KJ. Novel Insights into How Overnutrition Disrupts the Hypothalamic Actions of Leptin. Front Endocrinol (Lausanne) 2018; 9:89. [PMID: 29632515 PMCID: PMC5879088 DOI: 10.3389/fendo.2018.00089] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 02/23/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity has become a worldwide health problem, but we still do not understand the molecular mechanisms that contribute to overeating and low expenditure of energy. Leptin has emerged as a major regulator of energy balance through its actions in the hypothalamus. Importantly, obese people exhibit high circulating levels of leptin, yet the hypothalamus no longer responds normally to this hormone to suppress appetite or to increase energy expenditure. Several well-known hypotheses have been proposed to explain impaired central responsiveness to the effects of leptin in obesity, including defective transit across the blood-brain barrier at the arcuate nucleus, hypothalamic endoplasmic reticulum stress, maladaptive sterile inflammation in the hypothalamus, and overexpression of molecules that may inhibit leptin signaling. We also discuss a new explanation that is based on our group's recent discovery of a signaling pathway that we named "NSAPP" after its five main protein components. The NSAPP pathway consists of an oxide transport chain that causes a transient, targeted burst in intracellular hydrogen peroxide (H2O2) to inactivate redox-sensitive members of the protein tyrosine phosphatase gene family. The NSAPP oxide transport chain is required for full activation of canonical leptin signaling in neurons but fails to function normally in states of overnutrition. Remarkably, leptin and insulin both require the NSAPP oxide transport chain, suggesting that a defect in this pathway could explain simultaneous resistance to the appetite-suppressing effects of both hormones in obesity.
Collapse
Affiliation(s)
- Stefanie Fruhwürth
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Heike Vogel
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Annette Schürmann
- Department of Experimental Diabetology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München-Neuherberg, Germany
| | - Kevin Jon Williams
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medicine, Section of Endocrinology, Diabetes, and Metabolism, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- *Correspondence: Kevin Jon Williams,
| |
Collapse
|
321
|
Li C, Xu MM, Wang K, Adler AJ, Vella AT, Zhou B. Macrophage polarization and meta-inflammation. Transl Res 2018; 191:29-44. [PMID: 29154757 PMCID: PMC5776711 DOI: 10.1016/j.trsl.2017.10.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/14/2022]
Abstract
Chronic overnutrition and obesity induces low-grade inflammation throughout the body. Termed "meta-inflammation," this chronic state of inflammation is mediated by macrophages located within the colon, liver, muscle, and adipose tissue. A sentinel orchestrator of immune activity and homeostasis, macrophages adopt variable states of activation as a function of time and environmental cues. Meta-inflammation phenotypically skews these polarization states and has been linked to numerous metabolic disorders. The past decade has revealed several key regulators of macrophage polarization, including the signal transducer and activator of transcription family, the peroxisome proliferator-activated receptor gamma, the CCAAT-enhancer-binding proteins (C/EBP) family, and the interferon regulatory factors. Recent studies have also suggested that microRNAs and long noncoding RNA influence macrophage polarization. The pathogenic alteration of macrophage polarization in meta-inflammation is regulated by both extracellular and intracellular cues, resulting in distinct secretome profiles. Meta-inflammation-altered macrophage polarization has been linked to insulin insensitivity, atherosclerosis, inflammatory bowel disease, cancer, and autoimmunity. Thus, further mechanistic exploration into the skewing of macrophage polarization promises to have profound impacts on improving global health.
Collapse
Affiliation(s)
- Chuan Li
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Maria M Xu
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Kepeng Wang
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Adam J Adler
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn
| | - Anthony T Vella
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| | - Beiyan Zhou
- Department of Immunology, University of Connecticut, School of Medicine, Farmington, Conn.
| |
Collapse
|
322
|
Lynes MD, Tseng YH. Deciphering adipose tissue heterogeneity. Ann N Y Acad Sci 2018; 1411:5-20. [PMID: 28763833 PMCID: PMC5788721 DOI: 10.1111/nyas.13398] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/02/2017] [Accepted: 05/05/2017] [Indexed: 02/06/2023]
Abstract
Obesity is an excess accumulation of adipose tissue mass, and, together with its sequelae, in particular type II diabetes and metabolic syndrome, obesity presents a major health crisis. Although obesity is simply caused by increased adipose mass, the heterogeneity of adipose tissue in humans means that the response to increased energy balance is highly complex. Individual subjects with similar phenotypes may respond very differently to the same treatments; therefore, obesity may benefit from a personalized precision medicine approach. The variability in the development of obesity is indeed driven by differences in sex, genetics, and environment, but also by the various types of adipose tissue as well as the different cell types that compose it. By describing the distinct cell populations that reside in different fat depots, we can interpret the complex effect of these various players in the maintenance of whole-body energy homeostasis. To further understand adipose tissue, adipogenic differentiation and the transcriptional program of lipid accumulation must be investigated. As the cell- and depot-specific functions are described, they can be placed in the context of energy excess to understand how the heterogeneity of adipose tissue shapes individual metabolic status and condition.
Collapse
Affiliation(s)
- Matthew D Lynes
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts and Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts
| |
Collapse
|
323
|
Cummings NE, Williams EM, Kasza I, Konon EN, Schaid MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola Apelo SI, Cottrell SE, Geiger G, Barnes ME, Wisinski JA, Fenske RJ, Matkowskyj KA, Kimple ME, Alexander CM, Merrins MJ, Lamming DW. Restoration of metabolic health by decreased consumption of branched-chain amino acids. J Physiol 2017; 596:623-645. [PMID: 29266268 DOI: 10.1113/jp275075] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 11/20/2017] [Indexed: 12/20/2022] Open
Abstract
KEY POINTS We recently found that feeding healthy mice a diet with reduced levels of branched-chain amino acids (BCAAs), which are associated with insulin resistance in both humans and rodents, modestly improves glucose tolerance and slows fat mass gain. In the present study, we show that a reduced BCAA diet promotes rapid fat mass loss without calorie restriction in obese mice. Selective reduction of dietary BCAAs also restores glucose tolerance and insulin sensitivity to obese mice, even as they continue to consume a high-fat, high-sugar diet. A low BCAA diet transiently induces FGF21 (fibroblast growth factor 21) and increases energy expenditure. We suggest that dietary protein quality (i.e. the precise macronutrient composition of dietary protein) may impact the effectiveness of weight loss diets. ABSTRACT Obesity and diabetes are increasing problems around the world, and although even moderate weight loss can improve metabolic health, reduced calorie diets are notoriously difficult to sustain. Branched-chain amino acids (BCAAs; leucine, isoleucine and valine) are elevated in the blood of obese, insulin-resistant humans and rodents. We recently demonstrated that specifically reducing dietary levels of BCAAs has beneficial effects on the metabolic health of young, growing mice, improving glucose tolerance and modestly slowing fat mass gain. In the present study, we examine the hypothesis that reducing dietary BCAAs will promote weight loss, reduce adiposity, and improve blood glucose control in diet-induced obese mice with pre-existing metabolic syndrome. We find that specifically reducing dietary BCAAs rapidly reverses diet-induced obesity and improves glucoregulatory control in diet-induced obese mice. Most dramatically, mice eating an otherwise unhealthy high-calorie, high-sugar Western diet with reduced levels of BCAAs lost weight and fat mass rapidly until regaining a normal weight. Importantly, this normalization of weight was mediated not by caloric restriction or increased activity, but by increased energy expenditure, and was accompanied by a transient induction of the energy balance regulating hormone FGF21 (fibroblast growth factor 21). Consumption of a Western diet reduced in BCAAs was also accompanied by a dramatic improvement in glucose tolerance and insulin resistance. Our results link dietary BCAAs with the regulation of metabolic health and energy balance in obese animals, and suggest that specifically reducing dietary BCAAs may represent a highly translatable option for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- Nicole E Cummings
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth M Williams
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Ildiko Kasza
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Elizabeth N Konon
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Michael D Schaid
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Brian A Schmidt
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Chetan Poudel
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Dawn S Sherman
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Deyang Yu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Sebastian I Arriola Apelo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Dairy Science, University of Wisconsin-Madison, Madison, WI, USA
| | - Sara E Cottrell
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Rural and Urban Scholars in Community Health Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gabriella Geiger
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Macy E Barnes
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Jaclyn A Wisinski
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Rachel J Fenske
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Kristina A Matkowskyj
- William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA.,Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Michelle E Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, USA
| | - Caroline M Alexander
- McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew J Merrins
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Dudley W Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA.,William S. Middleton Memorial Veterans Hospital, Madison, WI, USA.,Endocrinology and Reproductive Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI, USA.,Interdisciplinary Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.,Molecular and Environmental Toxicology Program, University of Wisconsin-Madison, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
324
|
Wang Y, Paulo E, Wu D, Wu Y, Huang W, Chawla A, Wang B. Adipocyte Liver Kinase b1 Suppresses Beige Adipocyte Renaissance Through Class IIa Histone Deacetylase 4. Diabetes 2017; 66:2952-2963. [PMID: 28882900 PMCID: PMC5697944 DOI: 10.2337/db17-0296] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/30/2017] [Indexed: 12/18/2022]
Abstract
Uncoupling protein 1+ beige adipocytes are dynamically regulated by environment in rodents and humans; cold induces formation of beige adipocytes, whereas warm temperature and nutrient excess lead to their disappearance. Beige adipocytes can form through de novo adipogenesis; however, how "beiging" characteristics are maintained afterward is largely unknown. In this study, we show that beige adipocytes formed postnatally in subcutaneous inguinal white adipose tissue lost thermogenic gene expression and multilocular morphology at the adult stage, but cold restored their beiging characteristics, a phenomenon termed beige adipocyte renaissance. Ablation of these postnatal beige adipocytes inhibited cold-induced beige adipocyte formation in adult mice. Furthermore, we demonstrated that beige adipocyte renaissance was governed by liver kinase b1 and histone deacetylase 4 in white adipocytes. Although neither presence nor thermogenic function of uncoupling protein 1+ beige adipocytes contributed to metabolic fitness in adipocyte liver kinase b1-deficient mice, our results reveal an unexpected role of white adipocytes in maintaining properties of preexisting beige adipocytes.
Collapse
Affiliation(s)
- Yangmeng Wang
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA
| | - Esther Paulo
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Dongmei Wu
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Yixuan Wu
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Beckman Research Institute of City of Hope, Duarte, CA
| | - Ajay Chawla
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| | - Biao Wang
- Department of Physiology, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA
| |
Collapse
|
325
|
Villarroya F, Gavaldà-Navarro A, Peyrou M, Villarroya J, Giralt M. The Lives and Times of Brown Adipokines. Trends Endocrinol Metab 2017; 28:855-867. [PMID: 29113711 DOI: 10.1016/j.tem.2017.10.005] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022]
Abstract
Brown adipose tissue (BAT) is responsible for adaptive non-shivering thermogenesis. Moreover, brown fat secretes regulatory factors, so-called brown adipokines, that have autocrine, paracrine, and endocrine actions. Brown adipokines are either polypeptides or nonpeptidic molecules including lipid molecules and microRNAs. The secretory properties of brown fat are essential for tissue remodeling adaptations to thermogenic necessities. The endocrine properties of brown adipokines are thought to contribute to the association between BAT activity and a healthy metabolic profile in relation to glucose and lipid homeostasis. The identification and characterization of brown adipokines may allow the discovery of circulating biomarkers of BAT activity in humans, and will lead to the development of candidate tools for therapeutic interventions in metabolic diseases.
Collapse
Affiliation(s)
- Francesc Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain.
| | - Aleix Gavaldà-Navarro
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Marion Peyrou
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| | - Joan Villarroya
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Institut de Recerca Hospital de la Santa Creu i Sant Pau, Barcelona, Catalonia, Spain
| | - Marta Giralt
- Departament de Bioquímica i Biomedicina Molecular, Institut de Biomedicina (IBUB), Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red 'Fisiopatologia de la Obesidad y Nutrición', Madrid, Spain; Institut de Recerca Sant Joan de Déu, Barcelona, Catalonia, Spain
| |
Collapse
|
326
|
Ikeda K, Kang Q, Yoneshiro T, Camporez JP, Maki H, Homma M, Shinoda K, Chen Y, Lu X, Maretich P, Tajima K, Ajuwon KM, Soga T, Kajimura S. UCP1-independent signaling involving SERCA2b-mediated calcium cycling regulates beige fat thermogenesis and systemic glucose homeostasis. Nat Med 2017; 23:1454-1465. [PMID: 29131158 PMCID: PMC5727902 DOI: 10.1038/nm.4429] [Citation(s) in RCA: 452] [Impact Index Per Article: 56.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
Uncoupling protein 1 (UCP1) plays a central role in nonshivering thermogenesis in brown fat; however, its role in beige fat remains unclear. Here we report a robust UCP1-independent thermogenic mechanism in beige fat that involves enhanced ATP-dependent Ca2+ cycling by sarco/endoplasmic reticulum Ca2+-ATPase 2b (SERCA2b) and ryanodine receptor 2 (RyR2). Inhibition of SERCA2b impairs UCP1-independent beige fat thermogenesis in humans and mice as well as in pigs, a species that lacks a functional UCP1 protein. Conversely, enhanced Ca2+ cycling by activation of α1- and/or β3-adrenergic receptors or the SERCA2b-RyR2 pathway stimulates UCP1-independent thermogenesis in beige adipocytes. In the absence of UCP1, beige fat dynamically expends glucose through enhanced glycolysis, tricarboxylic acid metabolism and pyruvate dehydrogenase activity for ATP-dependent thermogenesis through the SERCA2b pathway; beige fat thereby functions as a 'glucose sink' and improves glucose tolerance independently of body weight loss. Our study uncovers a noncanonical thermogenic mechanism through which beige fat controls whole-body energy homeostasis via Ca2+ cycling.
Collapse
Affiliation(s)
- Kenji Ikeda
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Qianqian Kang
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Takeshi Yoneshiro
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Joao Paulo Camporez
- Yale University School of Medicine, Department of Medicine and Cellular & Molecular Physiology, New Haven, CT
| | - Hiroko Maki
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Mayu Homma
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Kosaku Shinoda
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Yong Chen
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Xiaodan Lu
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Pema Maretich
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Kazuki Tajima
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| | - Kolapo M. Ajuwon
- Department of Animal Sciences, Purdue University, West Lafayette, IN
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Shingo Kajimura
- UCSF Diabetes Center, San Francisco, CA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA
- Department of Cell and Tissue Biology, University of California, San Francisco, CA
| |
Collapse
|
327
|
Tsiloulis T, Carey AL, Bayliss J, Canny B, Meex RCR, Watt MJ. No evidence of white adipocyte browning after endurance exercise training in obese men. Int J Obes (Lond) 2017; 42:721-727. [PMID: 29188818 DOI: 10.1038/ijo.2017.295] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 10/31/2017] [Accepted: 11/10/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND/OBJECTIVES The phenomenon of adipocyte 'beiging' involves the conversion of non-classic brown adipocytes to brown-like adipose tissue with thermogenic, fat-burning properties, and this phenomenon has been shown in rodents to slow the progression of obesity-associated metabolic diseases. Rodent studies consistently report adipocyte beiging after endurance exercise training, indicating that increased thermogenic capacity in these adipocytes may underpin the improved health benefits of exercise training. The aim of this study was to determine whether prolonged endurance exercise training induces beige adipogenesis in subcutaneous adipose tissues of obese men. SUBJECTS/METHODS Molecular markers of beiging were examined in adipocytes obtained from abdominal subcutaneous (AbSC) and gluteofemoral (GF) subcutaneous adipose tissues before and after 6 weeks of endurance exercise training in obese men (n=6, 37.3±2.3 years, 30.1±2.3 kg m-2). RESULTS The mRNAs encoding the brown or beige adipocyte-selective proteins were very lowly expressed in AbSC and GF adipose tissues and exercise training did not alter the mRNA expression of UCP1, CD137, CITED, TBX1, LHX8 and TCF21. Using immunohistochemistry, neither multilocular adipocytes, nor UCP1 or CD137-positive adipocytes were detected in any sample. MicroRNAs known to regulate brown and/or beige adipose development were highly expressed in white adipocytes but endurance exercise training did not impact their expression. CONCLUSIONS The present study reaffirms emerging data in humans demonstrating no evidence of white adipose tissue beiging in response to exercise training, and supports a growing body of work demonstrating divergence of brown/beige adipose location, molecular characterization and physiological function between rodents and humans.
Collapse
Affiliation(s)
- T Tsiloulis
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and the Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - A L Carey
- Metabolic and Vascular Physiology Laboratory, Baker IDI Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - J Bayliss
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and the Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - B Canny
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and the Department of Physiology, Monash University, Clayton, Victoria, Australia.,School of Medicine, Faculty of Health, University of Tasmania, Hobart, Tasmania, Australia
| | - R C R Meex
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and the Department of Physiology, Monash University, Clayton, Victoria, Australia.,Department of Human Biology, School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - M J Watt
- Monash Biomedicine Discovery Institute, Metabolic Disease and Obesity Program, and the Department of Physiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
328
|
Milet C, Bléher M, Allbright K, Orgeur M, Coulpier F, Duprez D, Havis E. Egr1 deficiency induces browning of inguinal subcutaneous white adipose tissue in mice. Sci Rep 2017; 7:16153. [PMID: 29170465 PMCID: PMC5701004 DOI: 10.1038/s41598-017-16543-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022] Open
Abstract
Beige adipocyte differentiation within white adipose tissue, referred to as browning, is seen as a possible mechanism for increasing energy expenditure. The molecular regulation underlying the thermogenic browning process has not been entirely elucidated. Here, we identify the zinc finger transcription factor EGR1 as a negative regulator of the beige fat program. Loss of Egr1 in mice promotes browning in the absence of external stimulation and leads to an increase of Ucp1 expression, which encodes the key thermogenic mitochondrial uncoupling protein-1. Moreover, EGR1 is recruited to the proximal region of the Ucp1 promoter in subcutaneous inguinal white adipose tissue. Transcriptomic analysis of subcutaneous inguinal white adipose tissue in the absence of Egr1 identifies the molecular signature of white adipocyte browning downstream of Egr1 deletion and highlights a concomitant increase of beige differentiation marker and a decrease in extracellular matrix gene expression. Conversely, Egr1 overexpression in mesenchymal stem cells decreases beige adipocyte differentiation, while increasing extracellular matrix production. These results reveal a role for Egr1 in blocking energy expenditure via direct Ucp1 transcription repression and highlight Egr1 as a therapeutic target for counteracting obesity.
Collapse
Affiliation(s)
- Cécile Milet
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, Inserm U1156, IBPS-Developmental Biology Laboratory, F-75005, Paris, France
| | - Marianne Bléher
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, Inserm U1156, IBPS-Developmental Biology Laboratory, F-75005, Paris, France
| | | | - Mickael Orgeur
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, Inserm U1156, IBPS-Developmental Biology Laboratory, F-75005, Paris, France
| | - Fanny Coulpier
- École normale supérieure, PSL Research University, CNRS, Inserm, Institut de Biologie de l'École normale supérieure (IBENS), Plateforme Génomique, 75005, Paris, France
| | - Delphine Duprez
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, Inserm U1156, IBPS-Developmental Biology Laboratory, F-75005, Paris, France.
| | - Emmanuelle Havis
- Sorbonne Universités, UPMC Univ Paris 06, CNRS UMR7622, Inserm U1156, IBPS-Developmental Biology Laboratory, F-75005, Paris, France.
| |
Collapse
|
329
|
Shabalina IG, Vyssokikh MY, Gibanova N, Csikasz RI, Edgar D, Hallden-Waldemarson A, Rozhdestvenskaya Z, Bakeeva LE, Vays VB, Pustovidko AV, Skulachev MV, Cannon B, Skulachev VP, Nedergaard J. Improved health-span and lifespan in mtDNA mutator mice treated with the mitochondrially targeted antioxidant SkQ1. Aging (Albany NY) 2017; 9:315-339. [PMID: 28209927 PMCID: PMC5361666 DOI: 10.18632/aging.101174] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 02/03/2017] [Indexed: 11/25/2022]
Abstract
MtDNA mutator mice exhibit marked features of premature aging. We find that these mice treated from age of ≈100 days with the mitochondria-targeted antioxidant SkQ1 showed a delayed appearance of traits of aging such as kyphosis, alopecia, lowering of body temperature, body weight loss, as well as ameliorated heart, kidney and liver pathologies. These effects of SkQ1 are suggested to be related to an alleviation of the effects of an enhanced reactive oxygen species (ROS) level in mtDNA mutator mice: the increased mitochondrial ROS released due to mitochondrial mutations probably interact with polyunsaturated fatty acids in cardiolipin, releasing malondialdehyde and 4-hydroxynonenal that form protein adducts and thus diminishes mitochondrial functions. SkQ1 counteracts this as it scavenges mitochondrial ROS. As the results, the normal mitochondrial ultrastructure is preserved in liver and heart; the phosphorylation capacity of skeletal muscle mitochondria as well as the thermogenic capacity of brown adipose tissue is also improved. The SkQ1-treated mice live significantly longer (335 versus 290 days). These data may be relevant in relation to treatment of mitochondrial diseases particularly and the process of aging in general.
Collapse
Affiliation(s)
- Irina G Shabalina
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Mikhail Yu Vyssokikh
- The Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russian Federation
| | - Natalia Gibanova
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Robert I Csikasz
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Daniel Edgar
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden.,Present address: Buck Institute for research on aging, Novato, CA 94945, USA
| | - Anne Hallden-Waldemarson
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Zinaida Rozhdestvenskaya
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Lora E Bakeeva
- The Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russian Federation.,Institute of Mitoengineering, Moscow State University, 119992, Moscow, Russian Federation
| | - Valeria B Vays
- The Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russian Federation
| | - Antonina V Pustovidko
- The Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russian Federation
| | - Maxim V Skulachev
- Institute of Mitoengineering, Moscow State University, 119992, Moscow, Russian Federation
| | - Barbara Cannon
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Vladimir P Skulachev
- The Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992, Moscow, Russian Federation.,Institute of Mitoengineering, Moscow State University, 119992, Moscow, Russian Federation
| | - Jan Nedergaard
- The Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| |
Collapse
|
330
|
Acute and chronic cold exposure differentially affects the browning of porcine white adipose tissue. Animal 2017; 12:1435-1441. [PMID: 29143709 DOI: 10.1017/s1751731117002981] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Piglets are characteristically cold intolerant and thus susceptible to high mortality. However, browning of white adipose tissue (WAT) can induce non-shivering thermogenesis as a potential strategy to facilitate the animal's response to cold. Whether cold exposure can induce browning of subcutaneous WAT (sWAT) in piglets in a similar manner as it can in humans remains largely unknown. In this study, piglets were exposed to acute cold (4°C, 10 h) or chronic cold exposure (8°C, 15 days), and the genes and proteins of uncoupling protein 1 (UCP1)-dependent and independent thermogenesis, mitochondrial biogenesis, lipogenic and lipolytic processes were analysed. Interestingly, acute cold exposure induced browning of porcine sWAT, smaller adipocytes and the upregulated expression of UCP1, PGC1α, PGC1β, C/EBPβ, Cidea, UCP3, CKMT1 and PM20D1. Conversely, chronic cold exposure impaired the browning process, reduced mitochondrial numbers and the expression of browning markers, including UCP1, PGC1α and PRDM16. The present study demonstrated that acute cold exposure (but not chronic cold exposure) induces porcine sWAT browning. Thus, browning of porcine sWAT could be a novel strategy to balance the body temperature of piglets, and thus could be protective against cold exposure.
Collapse
|
331
|
Shin H, Ma Y, Chanturiya T, Cao Q, Wang Y, Kadegowda AKG, Jackson R, Rumore D, Xue B, Shi H, Gavrilova O, Yu L. Lipolysis in Brown Adipocytes Is Not Essential for Cold-Induced Thermogenesis in Mice. Cell Metab 2017; 26:764-777.e5. [PMID: 28988822 PMCID: PMC5905336 DOI: 10.1016/j.cmet.2017.09.002] [Citation(s) in RCA: 191] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 06/28/2017] [Accepted: 09/05/2017] [Indexed: 01/18/2023]
Abstract
Lipid droplet (LD) lipolysis in brown adipose tissue (BAT) is generally considered to be required for cold-induced nonshivering thermogenesis. Here, we show that mice lacking BAT Comparative Gene Identification-58 (CGI-58), a lipolytic activator essential for the stimulated LD lipolysis, have normal thermogenic capacity and are not cold sensitive. Relative to littermate controls, these animals had higher body temperatures when they were provided food during cold exposure. The increase in body temperature in the fed, cold-exposed knockout mice was associated with increased energy expenditure and with increased sympathetic innervation and browning of white adipose tissue (WAT). Mice lacking CGI-58 in both BAT and WAT were cold sensitive, but only in the fasted state. Thus, LD lipolysis in BAT is not essential for cold-induced nonshivering thermogenesis in vivo. Rather, CGI-58-dependent LD lipolysis in BAT regulates WAT thermogenesis, and our data uncover an essential role of WAT lipolysis in fueling thermogenesis during fasting.
Collapse
Affiliation(s)
- Hyunsu Shin
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; Center for Molecular and Translational Medicine, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Yinyan Ma
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; Mouse Metabolism Core Laboratory, The National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tatyana Chanturiya
- Mouse Metabolism Core Laboratory, The National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Qiang Cao
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA
| | - Youlin Wang
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; Center for Molecular and Translational Medicine, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA
| | - Anil K G Kadegowda
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Rachel Jackson
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Dominic Rumore
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Bingzhong Xue
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA
| | - Hang Shi
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30303, USA
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, The National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Liqing Yu
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA; Center for Molecular and Translational Medicine, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA 30303, USA.
| |
Collapse
|
332
|
Loh RKC, Kingwell BA, Carey AL. Human brown adipose tissue as a target for obesity management; beyond cold-induced thermogenesis. Obes Rev 2017; 18:1227-1242. [PMID: 28707455 DOI: 10.1111/obr.12584] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 06/01/2017] [Accepted: 06/01/2017] [Indexed: 02/01/2023]
Abstract
Elevating energy expenditure via adaptive thermogenesis in brown adipose tissue (BAT) is a potential strategy to reverse obesity. Much early enthusiasm for this approach, based on rodent studies, was tempered by the belief that BAT was relatively inconsequential in healthy adult humans. Interest was reinvigorated a decade ago when a series of studies re-identified BAT, primarily in upper thoracic regions, in adults. Despite the ensuing explosion of pre-clinical investigations and identification of an extensive list of potential target molecules for BAT recruitment, our understanding of human BAT physiology remains limited, particularly regarding interventions which might hold therapeutic promise. Cold-induced BAT thermogenesis (CIT) has been well studied, although is not readily translatable as an anti-obesity approach, whereas little is known regarding the role of BAT in human diet-induced thermogenesis (DIT). Furthermore, human studies dedicated to translating known pharmacological mechanisms of adipose browning from animal models are sparse. Several lines of recent evidence suggest that molecular regulation and physiology of human BAT differ to that of laboratory rodents, which form the majority of our knowledge base. This review will summarize knowledge on CIT and expand upon the current understanding and evidence gaps related to human adaptive thermogenesis via mechanisms other than cold.
Collapse
Affiliation(s)
- R K C Loh
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - B A Kingwell
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - A L Carey
- Metabolic and Vascular Physiology Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia
| |
Collapse
|
333
|
The thermogenic actions of natriuretic peptide in brown adipocytes: The direct measurement of the intracellular temperature using a fluorescent thermoprobe. Sci Rep 2017; 7:12978. [PMID: 29021616 PMCID: PMC5636787 DOI: 10.1038/s41598-017-13563-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022] Open
Abstract
In addition to the various effects of natriuretic peptides (NPs) on cardiovascular systems, increasing attention is being paid to the possibility that NPs induce adipose tissue browning and activate thermogenic program. We herein established a direct intracellular temperature measurement system using a fluorescent thermoprobe and investigated the thermogenic effects of A-type NP (ANP) on brown adipocytes. The thermoprobe was successfully introduced into rat brown adipocytes, and the temperature dependent change in fluorescence intensity ratio was measured using a fluorescence microscope. After one-hour incubation with ANP, the degree of the change in fluorescence intensity ratio was significantly higher in ANP-treated (P < 0.01) adipocytes compared to untreated controls. The ANP treatment increased uncoupling protein-1 (UCP1) mRNA levels, which is one of the markers of thermogenesis in adipocytes, while the intracellular ATP content was not changed, indicating mitochondrial uncoupled respiration. Intriguingly, these thermogenic actions of ANP were more prominent when brown adipocytes were incubated at 35 °C than at 37 °C. Moreover, the increase in the intracellular temperature and the expression of UCP1 induced by ANP were cancelled by p38MAPK inhibition. Taken together, this study directly demonstrated the thermogenic actions of ANP in brown adipocytes through the use of a novel method of intracellular temperature measurement.
Collapse
|
334
|
Okamatsu-Ogura Y, Nio-Kobayashi J, Nagaya K, Tsubota A, Kimura K. Brown adipocytes postnatally arise through both differentiation from progenitors and conversion from white adipocytes in Syrian hamster. J Appl Physiol (1985) 2017; 124:99-108. [PMID: 28982944 DOI: 10.1152/japplphysiol.00622.2017] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
To investigate the postnatal development of brown adipose tissue (BAT) in Syrian hamsters, we histologically examined interscapular fat tissue from 5-16-day-old pups, focusing on how brown adipocytes arise. Interscapular fat of 5-day-old hamsters mainly consisted of white adipocytes containing large unilocular lipid droplets, as observed in typical white adipose tissue (WAT). On day 7, clusters of small, proliferative nonadipocytes with a strong immunoreactivity for Ki67 appeared near the edge of the interscapular fat tissue. The area of the Ki67-positive regions expanded to ~50% of the total tissue area by day 10. The interscapular fat showed the typical BAT feature by day 16. A brown adipocyte-specific marker, uncoupling protein-1, was clearly detected on day 10 and thereafter, while not detected on day 7. During conversion of interscapular fat from WAT to BAT, unilocular adipocytes completely and rapidly disappeared without obvious apoptosis. Dual immunofluorescence staining for Ki67 and monocarboxylate transporter 1 (MCT1), another selective marker for brown adipocytes, revealed that most of the proliferating cells were of the brown adipocyte lineage. Electron microscopic examination showed that some of the white adipocytes contained small lipid droplets in addition to the large droplet and expressed MCT1 as do progenitor and mature brown adipocytes, implying a direct conversion from white to brown adipocytes. These results suggest that BAT of Syrian hamsters develops postnatally through two different pathways: the proliferation and differentiation of brown adipocyte progenitors and the conversion of unilocular adipocytes to multilocular brown adipocytes. NEW & NOTEWORTHY Brown and white adipose tissues (BAT and WAT, respectively) are quite different in morphological features and function; however, the boundary between these tissues is obscure. In this study, we histologically evaluated the process of BAT development in Syrian hamsters, which shows postnatal conversion of WAT to BAT. Our results suggest that brown adipocytes arise through two different pathways: the proliferation and differentiation of brown adipocyte progenitors and the conversion from white adipocytes.
Collapse
Affiliation(s)
- Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Junko Nio-Kobayashi
- Laboratory of Histology and Cytology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University , Sapporo , Japan
| | - Kazuki Nagaya
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Ayumi Tsubota
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| | - Kazuhiro Kimura
- Laboratory of Biochemistry, Division of Veterinary Medicine, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo , Japan
| |
Collapse
|
335
|
Li G, Xie C, Lu S, Nichols RG, Tian Y, Li L, Patel D, Ma Y, Brocker CN, Yan T, Krausz KW, Xiang R, Gavrilova O, Patterson AD, Gonzalez FJ. Intermittent Fasting Promotes White Adipose Browning and Decreases Obesity by Shaping the Gut Microbiota. Cell Metab 2017; 26:672-685.e4. [PMID: 28918936 PMCID: PMC5668683 DOI: 10.1016/j.cmet.2017.08.019] [Citation(s) in RCA: 418] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/10/2017] [Accepted: 08/17/2017] [Indexed: 12/20/2022]
Abstract
While activation of beige thermogenesis is a promising approach for treatment of obesity-associated diseases, there are currently no known pharmacological means of inducing beiging in humans. Intermittent fasting is an effective and natural strategy for weight control, but the mechanism for its efficacy is poorly understood. Here, we show that an every-other-day fasting (EODF) regimen selectively stimulates beige fat development within white adipose tissue and dramatically ameliorates obesity, insulin resistance, and hepatic steatosis. EODF treatment results in a shift in the gut microbiota composition leading to elevation of the fermentation products acetate and lactate and to the selective upregulation of monocarboxylate transporter 1 expression in beige cells. Microbiota-depleted mice are resistance to EODF-induced beiging, while transplantation of the microbiota from EODF-treated mice to microbiota-depleted mice activates beiging and improves metabolic homeostasis. These findings provide a new gut-microbiota-driven mechanism for activating adipose tissue browning and treating metabolic diseases.
Collapse
Affiliation(s)
- Guolin Li
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China; The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.
| | - Cen Xie
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Siyu Lu
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Robert G Nichols
- Department of Molecular Toxicology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Yuan Tian
- Department of Molecular Toxicology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Licen Li
- The Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Daxeshkumar Patel
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yinyan Ma
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chad N Brocker
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Tingting Yan
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kristopher W Krausz
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rong Xiang
- The State Key Laboratory of Medical Genetics & School of Life Sciences, Central South University, Changsha, Hunan 41001, China
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andrew D Patterson
- Department of Molecular Toxicology, The Pennsylvania State University, University Park, PA 16802, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
336
|
Systems biology reveals uncoupling beyond UCP1 in human white fat-derived beige adipocytes. NPJ Syst Biol Appl 2017; 3:29. [PMID: 28983409 PMCID: PMC5626775 DOI: 10.1038/s41540-017-0027-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 08/04/2017] [Accepted: 08/23/2017] [Indexed: 01/08/2023] Open
Abstract
Pharmaceutical induction of metabolically active beige adipocytes in the normally energy storing white adipose tissue has potential to reduce obesity. Mitochondrial uncoupling in beige adipocytes, as in brown adipocytes, has been reported to occur via the uncoupling protein 1 (UCP1). However, several previous in vitro characterizations of human beige adipocytes have only measured UCP1 mRNA fold increase, and assumed a direct correlation with metabolic activity. Here, we provide an example of pharmaceutical induction of beige adipocytes, where increased mRNA levels of UCP1 are not translated into increased protein levels, and perform a thorough analysis of this example. We incorporate mRNA and protein levels of UCP1, time-resolved mitochondrial characterizations, and numerous perturbations, and analyze all data with a new fit-for-purpose mathematical model. The systematic analysis challenges the seemingly obvious experimental conclusion, i.e., that UCP1 is not active in the induced cells, and shows that hypothesis testing with iterative modeling and experimental work is needed to sort out the role of UCP1. The analyses demonstrate, for the first time, that the uncoupling capability of human beige adipocytes can be obtained without UCP1 activity. This finding thus opens the door to a new direction in drug discovery that targets obesity and its associated comorbidities. Furthermore, the analysis advances our understanding of how to evaluate UCP1-independent thermogenesis in human beige adipocytes.
Collapse
|
337
|
van den Berg SM, van Dam AD, Kusters PJH, Beckers L, den Toom M, van der Velden S, Van den Bossche J, van Die I, Boon MR, Rensen PCN, Lutgens E, de Winther MPJ. Helminth antigens counteract a rapid high-fat diet-induced decrease in adipose tissue eosinophils. J Mol Endocrinol 2017; 59:245-255. [PMID: 28694301 DOI: 10.1530/jme-17-0112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 07/10/2017] [Indexed: 12/16/2022]
Abstract
Brown adipose tissue (BAT) activation and white adipose tissue (WAT) beiging can increase energy expenditure and have the potential to reduce obesity and associated diseases. The immune system is a potential target in mediating brown and beige adipocyte activation. Type 2 and anti-inflammatory immune cells contribute to metabolic homeostasis within lean WAT, with a prominent role for eosinophils and interleukin (IL)-4-induced anti-inflammatory macrophages. We determined eosinophil numbers in epididymal WAT (EpAT), subcutaneous WAT (ScAT) and BAT after 1 day, 3 days or 1 week of high-fat diet (HFD) feeding in C57Bl/6 mice. One day of HFD resulted in a rapid drop in eosinophil numbers in EpAT and BAT, and after 3 days, in ScAT. In an attempt to restore this HFD-induced drop in adipose tissue eosinophils, we treated 1-week HFD-fed mice with helminth antigens from Schistosoma mansoni or Trichuris suis and evaluated whether the well-known protective metabolic effects of helminth antigens involves BAT activation or beiging. Indeed, antigens of both helminth species induced high numbers of eosinophils in EpAT, but failed to induce beiging. In ScAT, Schistosoma mansoni antigens induced mild eosinophilia, which was accompanied by slightly more beiging. No effects were observed in BAT. To study type 2 responses on brown adipocytes directly, T37i cells were stimulated with IL-4. This increased Ucp1 expression and strongly induced the production of eosinophil chemoattractant CCL11 (+26-fold), revealing that brown adipocytes themselves can attract eosinophils. Our findings indicate that helminth antigen-induced eosinophilia fails to induce profound beiging of white adipocytes.
Collapse
Affiliation(s)
- Susan M van den Berg
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Andrea D van Dam
- Department of MedicineDivision Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Center, Leiden, The Netherlands
| | - Pascal J H Kusters
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Linda Beckers
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Myrthe den Toom
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Saskia van der Velden
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan Van den Bossche
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Irma van Die
- Department of Molecular Cell Biology and ImmunologyVU University Medical Center, Amsterdam, The Netherlands
| | - Mariëtte R Boon
- Department of MedicineDivision Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Center, Leiden, The Netherlands
| | - Patrick C N Rensen
- Department of MedicineDivision Endocrinology, Leiden University Medical Center, Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical Center, Leiden, The Netherlands
| | - Esther Lutgens
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK)Ludwig Maximilian's University, Munich, Germany
| | - Menno P J de Winther
- Department of Medical BiochemistryExperimental Vascular Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Institute for Cardiovascular Prevention (IPEK)Ludwig Maximilian's University, Munich, Germany
| |
Collapse
|
338
|
Markussen LK, Isidor MS, Breining P, Andersen ES, Rasmussen NE, Petersen LI, Pedersen SB, Richelsen B, Hansen JB. Characterization of immortalized human brown and white pre-adipocyte cell models from a single donor. PLoS One 2017; 12:e0185624. [PMID: 28957413 PMCID: PMC5619805 DOI: 10.1371/journal.pone.0185624] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 09/15/2017] [Indexed: 12/22/2022] Open
Abstract
Brown adipose tissue with its constituent brown adipocytes is a promising therapeutic target in metabolic disorders due to its ability to dissipate energy and improve systemic insulin sensitivity and glucose homeostasis. The molecular control of brown adipocyte differentiation and function has been extensively studied in mice, but relatively little is known about such regulatory mechanisms in humans, which in part is due to lack of human brown adipose tissue derived cell models. Here, we used retrovirus-mediated overexpression to stably integrate human telomerase reverse transcriptase (TERT) into stromal-vascular cell fractions from deep and superficial human neck adipose tissue biopsies from the same donor. The brown and white pre-adipocyte cell models (TERT-hBA and TERT-hWA, respectively) displayed a stable proliferation rate and differentiation until at least passage 20. Mature TERT-hBA adipocytes expressed higher levels of thermogenic marker genes and displayed a higher maximal respiratory capacity than mature TERT-hWA adipocytes. TERT-hBA adipocytes were UCP1-positive and responded to β-adrenergic stimulation by activating the PKA-MKK3/6-p38 MAPK signaling module and increasing thermogenic gene expression and oxygen consumption. Mature TERT-hWA adipocytes underwent efficient rosiglitazone-induced ‘browning’, as demonstrated by strongly increased expression of UCP1 and other brown adipocyte-enriched genes. In summary, the TERT-hBA and TERT-hWA cell models represent useful tools to obtain a better understanding of the molecular control of human brown and white adipocyte differentiation and function as well as of browning of human white adipocytes.
Collapse
Affiliation(s)
| | - Marie S. Isidor
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Peter Breining
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Elise S. Andersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | | | - Steen B. Pedersen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Bjørn Richelsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jacob B. Hansen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
339
|
Kim HL, Jung Y, Park J, Youn DH, Kang J, Lim S, Lee BS, Jeong MY, Choe SK, Park R, Ahn KS, Um JY. Farnesol Has an Anti-obesity Effect in High-Fat Diet-Induced Obese Mice and Induces the Development of Beige Adipocytes in Human Adipose Tissue Derived-Mesenchymal Stem Cells. Front Pharmacol 2017; 8:654. [PMID: 29033835 PMCID: PMC5627035 DOI: 10.3389/fphar.2017.00654] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/01/2017] [Indexed: 12/17/2022] Open
Abstract
Brown adipocytes dissipate energy as heat and hence have an important therapeutic capacity for obesity. Development of brown-like adipocytes (also called beige) is also another attractive target for obesity treatment. Here, we investigated the effect of farnesol, an isoprenoid, on adipogenesis in adipocytes and on the browning of white adipose tissue (WAT) as well as on the weight gain of high-fat diet (HFD)-induced obese mice. Farnesol inhibited adipogenesis and the related key regulators including peroxisome proliferator-activated receptor γ (PPARγ) and CCAAT/enhancer binding protein α through the up-regulation of AMP-activated protein kinase in 3T3-L1 murine adipocytes and human adipose tissue-derived mesenchymal stem cells (hAMSCs). Farnesol markedly increased the expression of uncoupling protein 1 and PPARγ coactivator 1 α in differentiated hAMSCs. In addition, farnesol limited the weight gain in HFD obese mice and induced the development of beige adipocytes in both inguinal and epididymal WAT. These results suggest that farnesol could be a potential therapeutic agent for obesity treatment.
Collapse
Affiliation(s)
- Hye-Lin Kim
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Yunu Jung
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Jinbong Park
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Dong-Hyun Youn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - JongWook Kang
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea.,Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Seona Lim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Beom Su Lee
- Department of Science in Korean Medicine, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Mi-Young Jeong
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Seong-Kyu Choe
- Department of Microbiology and Center for Metabolic Function Regulation, School of Medicine, Wonkwang UniversityIksan, South Korea
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and TechnologyGwangju, South Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| | - Jae-Young Um
- College of Korean Medicine, Basic Research Laboratory for Comorbidity Regulation, Graduate School, Kyung Hee UniversitySeoul, South Korea
| |
Collapse
|
340
|
Ying F, Cai Y, Cai Y, Wang Y, Ching Tang EH. Prostaglandin E receptor subtype 4 regulates lipid droplet size and mitochondrial activity in murine subcutaneous white adipose tissue. FASEB J 2017; 31:4023-4036. [DOI: 10.1096/fj.201700191r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Fan Ying
- Department of Pharmacology and PharmacyState Key Laboratory of Pharmaceutical BiotechnologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yin Cai
- Department of AnesthesiologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yu Cai
- Department of Pharmacology and PharmacyState Key Laboratory of Pharmaceutical BiotechnologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Yu Wang
- Department of Pharmacology and PharmacyState Key Laboratory of Pharmaceutical BiotechnologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Eva Hoi Ching Tang
- Department of Pharmacology and PharmacyState Key Laboratory of Pharmaceutical BiotechnologyLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
- School of Biomedical SciencesLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| |
Collapse
|
341
|
Zou Y, Lu P, Shi J, Liu W, Yang M, Zhao S, Chen N, Chen M, Sun Y, Gao A, Chen Q, Zhang Z, Ma Q, Ning T, Ying X, Jin J, Deng X, Shen B, Zhang Y, Yuan B, Kauderer S, Liu S, Hong J, Liu R, Ning G, Wang W, Gu W, Wang J. IRX3 Promotes the Browning of White Adipocytes and Its Rare Variants are Associated with Human Obesity Risk. EBioMedicine 2017; 24:64-75. [PMID: 28988979 PMCID: PMC5652024 DOI: 10.1016/j.ebiom.2017.09.010] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 12/31/2022] Open
Abstract
Background IRX3 was recently reported as the effector of the FTO variants. We aimed to test IRX3's roles in the browning program and to evaluate the association between the genetic variants in IRX3 and human obesity. Methods IRX3 expression was examined in beige adipocytes in human and mouse models, and further validated in induced beige adipocytes. The browning capacity of primary preadipocytes was assessed with IRX3 knockdown. Luciferase reporter analysis and ChIP assay were applied to investigate IRX3's effects on UCP1 transcriptional activity. Moreover, genetic analysis of IRX3 was performed in 861 young obese subjects and 916 controls. Results IRX3 expression was induced in the browning process and was positively correlated with the browning markers. IRX3 knockdown remarkably inhibited UCP1 expression in induced mouse and human beige adipocytes, and also repressed the uncoupled oxygen consumption rate. Further, IRX3 directly bound to UCP1 promoter and increased its transcriptional activity. Moreover, 17 rare heterozygous missense/frameshift IRX3 variants were identified, with a significant enrichment in obese subjects (P = 0.038, OR = 2.27; 95% CI, 1.02–5.05). Conclusions IRX3 deficiency repressed the browning program of white adipocytes partially by regulating UCP1 transcriptional activity. Rare variants of IRX3 were associated with human obesity. IRX3 expression was positively correlated with the browning markers in both mice and humans. IRX3 deficiency repressed the browning program in human and mouse cell models by regulating UCP1 transcriptional activity. Rare variants of IRX3 were associated with human obesity.
While FTO is the first risk gene for obesity identified via genome-wide association studies, its exact mechanism remains unknown until two recent studies reported that IRX3, as FTO's target, probably inhibited the browning of white fat. However, whether IRX3 gene per se predisposes to obesity in humans and how IRX3 regulates the browning process remains unclear. Herein we combined cellular and genetic evidence to support that IRX3 promotes but not inhibit the browning process in both human and mouse cell models. Mechanistically, IRX3 increased the transcriptional activity of UCP1 through directly binding to its promoter. Thus, our findings provided an intact picture of IRX3's function in browning program and human obesity.
Collapse
Affiliation(s)
- Yaoyu Zou
- Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China; Shanghai Ji Ai Genetics & IVF Institute, Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Peng Lu
- Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Juan Shi
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Wen Liu
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Minglan Yang
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Shaoqian Zhao
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Na Chen
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Maopei Chen
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Yingkai Sun
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Aibo Gao
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Qingbo Chen
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Zhiguo Zhang
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Qinyun Ma
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Tinglu Ning
- Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xiayang Ying
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Jiabin Jin
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Xiaxing Deng
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Baiyong Shen
- Pancreatic Disease Center, Department of General Surgery, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Yifei Zhang
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Bo Yuan
- Department of Burns and Plastic Surgery, Ruijin Hospital, SJTUSM, Shanghai 200025, China
| | - Sophie Kauderer
- Department of Epidemiology and Center for Global Cardiometabolic Health, School of Public Health, Department of Medicine (Endocrinology), Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Simin Liu
- Department of Epidemiology and Center for Global Cardiometabolic Health, School of Public Health, Department of Medicine (Endocrinology), Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Jie Hong
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China
| | - Guang Ning
- Shanghai Institutes for Biological Sciences, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China.
| | - Weiqing Wang
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China.
| | - Weiqiong Gu
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China.
| | - Jiqiu Wang
- Department of Endocrinology and Metabolism, National Key Laboratory for Medical Genomes, China National Research Center for Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 200025, China.
| |
Collapse
|
342
|
Castro É, Silva TEO, Festuccia WT. Critical review of beige adipocyte thermogenic activation and contribution to whole-body energy expenditure. Horm Mol Biol Clin Investig 2017; 31:/j/hmbci.ahead-of-print/hmbci-2017-0042/hmbci-2017-0042.xml. [PMID: 28862985 DOI: 10.1515/hmbci-2017-0042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 07/17/2017] [Indexed: 12/30/2022]
Abstract
Beige (or brite, "brown in white") adipocytes are uncoupling protein 1 (UCP1)-positive cells residing in white adipose depots that, depending on the conditions, behave either as classic white adipocytes, storing energy as lipids, or as brown adipocytes, dissipating energy from oxidative metabolism as heat through non-shivering thermogenesis. Because of their thermogenic potential and, therefore, possible usage to treat metabolic diseases such as obesity and type 2 diabetes, beige cells have attracted the attention of many scientists worldwide aiming to develop strategies to safely recruit and activate their thermogenic activity. Indeed, in recent years, a large variety of conditions, molecules (including nutrients) and signaling pathways were reported to promote the recruitment of beige adipocytes. Despite of those advances, the true contribution of beige adipocyte thermogenesis to whole-body energy expenditure is still not completely defined. Herein, we discuss some important aspects that should be considered when studying beige adipocyte biology and the contribution to energy balance and whole-body metabolism.
Collapse
|
343
|
Chouchani ET, Kazak L, Spiegelman BM. Mitochondrial reactive oxygen species and adipose tissue thermogenesis: Bridging physiology and mechanisms. J Biol Chem 2017; 292:16810-16816. [PMID: 28842500 DOI: 10.1074/jbc.r117.789628] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Brown and beige adipose tissues can catabolize stored energy to generate heat, relying on the principal effector of thermogenesis: uncoupling protein 1 (UCP1). This unique capability could be leveraged as a therapy for metabolic disease. Numerous animal and cellular models have now demonstrated that mitochondrial reactive oxygen species (ROS) signal to support adipocyte thermogenic identity and function. Herein, we contextualize these findings within the established principles of redox signaling and mechanistic studies of UCP1 function. We provide a framework for understanding the role of mitochondrial ROS signaling in thermogenesis together with testable hypotheses for understanding mechanisms and developing therapies.
Collapse
Affiliation(s)
- Edward T Chouchani
- From the Dana-Farber Cancer Institute, Harvard Medical School and.,Department of Cell Biology, Harvard University Medical School, Boston, Massachusetts 02115
| | - Lawrence Kazak
- From the Dana-Farber Cancer Institute, Harvard Medical School and.,Department of Cell Biology, Harvard University Medical School, Boston, Massachusetts 02115
| | - Bruce M Spiegelman
- From the Dana-Farber Cancer Institute, Harvard Medical School and .,Department of Cell Biology, Harvard University Medical School, Boston, Massachusetts 02115
| |
Collapse
|
344
|
Kreisler A, Garcia M, Spierling S, Hui B, Zorrilla E. Extended vs. brief intermittent access to palatable food differently promote binge-like intake, rejection of less preferred food, and weight cycling in female rats. Physiol Behav 2017; 177:305-316. [DOI: 10.1016/j.physbeh.2017.03.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/08/2017] [Accepted: 03/27/2017] [Indexed: 01/16/2023]
|
345
|
Browning deficiency and low mobilization of fatty acids in gonadal white adipose tissue leads to decreased cold-tolerance of transglutaminase 2 knock-out mice. Biochim Biophys Acta Mol Cell Biol Lipids 2017; 1862:1575-1586. [PMID: 28774822 DOI: 10.1016/j.bbalip.2017.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 07/03/2017] [Accepted: 07/28/2017] [Indexed: 12/12/2022]
Abstract
During cold-exposure 'beige' adipocytes with increased mitochondrial content are activated in white adipose tissue (WAT). These cells, similarly to brown adipose tissue (BAT), dissipate stored chemical energy in the form of heat with the help of uncoupling protein 1 (UCP1). We investigated the effect of tissue transglutaminase (TG2) ablation on the function of ATs in mice. Although TG2+/+ and TG2-/- mice had the same amount of WAT and BAT, we found that TG2+/+ animals could tolerate acute cold exposure for 4h, whereas TG2-/- mice only for 3h. Both TG2-/- and TG2+/+ animals used up half of the triacylglycerol content of subcutaneous WAT (SCAT) after 3h treatment; however, TG2-/- mice still possessed markedly whiter and higher amount of gonadal WAT (GONAT) as reflected in the larger size of adipocytes and lower free fatty acid levels in serum. Furthermore, lower expression of 'beige' marker genes such as UCP1, TBX1 and TNFRFS9 was observed after cold exposure in GONAT of TG2-/- mice, paralleled with a lower level of UCP1 protein and a decreased mitochondrial content. The detected changes in gene expression of Resistin and Adiponectin did not provoke glucose intolerance in the investigated TG2-/- mice, and TG2 deletion did not influence adrenaline, noradrenaline, glucagon and insulin production. Our data suggest that TG2 has a tissue-specific role in GONAT function and browning, which becomes apparent under acute cold exposure.
Collapse
|
346
|
Keipert S, Kutschke M, Ost M, Schwarzmayr T, van Schothorst EM, Lamp D, Brachthäuser L, Hamp I, Mazibuko SE, Hartwig S, Lehr S, Graf E, Plettenburg O, Neff F, Tschöp MH, Jastroch M. Long-Term Cold Adaptation Does Not Require FGF21 or UCP1. Cell Metab 2017; 26:437-446.e5. [PMID: 28768181 DOI: 10.1016/j.cmet.2017.07.016] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 05/31/2017] [Accepted: 07/18/2017] [Indexed: 11/13/2022]
Abstract
Brown adipose tissue (BAT)-dependent thermogenesis and its suggested augmenting hormone, FGF21, are potential therapeutic targets in current obesity and diabetes research. Here, we studied the role of UCP1 and FGF21 for metabolic homeostasis in the cold and dissected underlying molecular mechanisms using UCP1-FGF21 double-knockout mice. We report that neither UCP1 nor FGF21, nor even compensatory increases of FGF21 serum levels in UCP1 knockout mice, are required for defense of body temperature or for maintenance of energy metabolism and body weight. Remarkably, cold-induced browning of inguinal white adipose tissue (iWAT) is FGF21 independent. Global RNA sequencing reveals major changes in response to UCP1- but not FGF21-ablation in BAT, iWAT, and muscle. Markers of mitochondrial failure and inflammation are observed in BAT, but in particular the enhanced metabolic reprogramming in iWAT supports the thermogenic role of UCP1 and excludes an important thermogenic role of endogenous FGF21 in normal cold acclimation.
Collapse
Affiliation(s)
- Susanne Keipert
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Maria Kutschke
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Mario Ost
- German Institute of Human Nutrition, Nuthetal, Germany
| | - Thomas Schwarzmayr
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | - Daniel Lamp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Laura Brachthäuser
- Institute of Pathology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Isabel Hamp
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Sithandiwe E Mazibuko
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Sonja Hartwig
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Düsseldorf, Germany
| | - Stefan Lehr
- German Center for Diabetes Research (DZD), Neuherberg, Germany; Institute for Clinical Biochemistry and Pathobiochemistry, German Diabetes Center (DDZ), Düsseldorf, Germany
| | - Elisabeth Graf
- Institute of Human Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Oliver Plettenburg
- Institute of Medicinal Chemistry, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Frauke Neff
- Institute of Pathology, Helmholtz-Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Division of Metabolic Diseases, Department of Medicine, Technische Universität München, München, Germany
| | - Martin Jastroch
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Department of Animal Physiology, Faculty of Biology, Philipps University of Marburg, Marburg, Germany.
| |
Collapse
|
347
|
Dodd GT, Andrews ZB, Simonds SE, Michael NJ, DeVeer M, Brüning JC, Spanswick D, Cowley MA, Tiganis T. A Hypothalamic Phosphatase Switch Coordinates Energy Expenditure with Feeding. Cell Metab 2017; 26:375-393.e7. [PMID: 28768176 DOI: 10.1016/j.cmet.2017.07.013] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 04/13/2017] [Accepted: 07/17/2017] [Indexed: 12/17/2022]
Abstract
Beige adipocytes can interconvert between white and brown-like states and switch between energy storage versus expenditure. Here we report that beige adipocyte plasticity is important for feeding-associated changes in energy expenditure and is coordinated by the hypothalamus and the phosphatase TCPTP. A fasting-induced and glucocorticoid-mediated induction of TCPTP, inhibited insulin signaling in AgRP/NPY neurons, repressed the browning of white fat and decreased energy expenditure. Conversely feeding reduced hypothalamic TCPTP, to increase AgRP/NPY neuronal insulin signaling, white adipose tissue browning and energy expenditure. The feeding-induced repression of hypothalamic TCPTP was defective in obesity. Mice lacking TCPTP in AgRP/NPY neurons were resistant to diet-induced obesity and had increased beige fat activity and energy expenditure. The deletion of hypothalamic TCPTP in obesity restored feeding-induced browning and increased energy expenditure to promote weight loss. Our studies define a hypothalamic switch that coordinates energy expenditure with feeding for the maintenance of energy balance.
Collapse
Affiliation(s)
- Garron T Dodd
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia
| | - Zane B Andrews
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| | - Stephanie E Simonds
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| | - Natalie J Michael
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| | - Michael DeVeer
- Monash Biomedical Imaging, Monash University, Victoria 3168, Australia
| | - Jens C Brüning
- Max Plank Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes, and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Land Str. 1, 85764 Neuherberg, Germany
| | - David Spanswick
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| | - Michael A Cowley
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Physiology, Monash University, Victoria 3800, Australia
| | - Tony Tiganis
- Metabolic Disease and Obesity Program, Biomedicine Discovery Institute, Monash University, Victoria 3800, Australia; Department of Biochemistry and Molecular Biology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
348
|
Kim M, Furuzono T, Yamakuni K, Li Y, Kim YI, Takahashi H, Ohue-Kitano R, Jheng HF, Takahashi N, Kano Y, Yu R, Kishino S, Ogawa J, Uchida K, Yamazaki J, Tominaga M, Kawada T, Goto T. 10-oxo-12( Z)-octadecenoic acid, a linoleic acid metabolite produced by gut lactic acid bacteria, enhances energy metabolism by activation of TRPV1. FASEB J 2017; 31:5036-5048. [PMID: 28754711 DOI: 10.1096/fj.201700151r] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/17/2017] [Indexed: 11/11/2022]
Abstract
Gut microbiota can regulate the host energy metabolism; however, the underlying mechanisms that could involve gut microbiota-derived compounds remain to be understood. Therefore, in this study, we investigated the effects of KetoA [10-oxo-12(Z)-octadecenoic acid]-a linoleic acid metabolite produced by gut lactic acid bacteria-on whole-body energy metabolism and found that dietary intake of KetoA could enhance energy expenditure in mice, thereby protecting mice from diet-induced obesity. By using Ca2+ imaging and whole-cell patch-clamp methods, KetoA was noted to potently activate transient receptor potential vanilloid 1 (TRPV1) and enhance noradrenalin turnover in adipose tissues. In addition, KetoA up-regulated genes that are related to brown adipocyte functions, including uncoupling protein 1 (UCP1) in white adipose tissue (WAT), which was later diminished in the presence of a β-adrenoreceptor blocker. By using obese and diabetic model KK-Ay mice, we further show that KetoA intake ameliorated obesity-associated metabolic disorders. In the absence of any observed KetoA-induced antiobesity effect or UCP1 up-regulation in TRPV1-deficient mice, we prove that the antiobesity effect of KetoA was caused by TRPV1 activation-mediated browning in WAT. KetoA produced in the gut could therefore be involved in the regulation of host energy metabolism.-Kim, M., Furuzono, T., Yamakuni, K., Li, Y., Kim, Y.-I., Takahashi, H., Ohue-Kitano, R., Jheng, H.-F., Takahashi, N., Kano, Y., Yu, R., Kishino, S., Ogawa, J., Uchida, K., Yamazaki, J., Tominaga, M., Kawada, T., Goto, T. 10-oxo-12(Z)-octadecenoic acid, a linoleic acid metabolite produced by gut lactic acid bacteria, enhances energy metabolism by activation of TRPV1.
Collapse
Affiliation(s)
- Minji Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Tomoya Furuzono
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kanae Yamakuni
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Yongjia Li
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Young-Il Kim
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Haruya Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Ryuji Ohue-Kitano
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Huei-Fen Jheng
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Nobuyuki Takahashi
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Yuriko Kano
- Laboratory of Nutrition Chemistry, Faculty of Home Economics, Kobe Women's University, Kobe, Japan
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, South Korea
| | - Shigenobu Kishino
- Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Jun Ogawa
- Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan.,Laboratory of Fermentation Physiology and Applied Microbiology, Division of Applied Life Sciences, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
| | - Kunitoshi Uchida
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Japan.,Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Jun Yamazaki
- Department of Physiological Science and Molecular Biology, Fukuoka Dental College, Fukuoka, Japan
| | - Makoto Tominaga
- Division of Cell Signaling, Okazaki Institute for Integrative Bioscience, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Japan.,Department of Physiological Sciences, The Graduate University for Advanced Studies, Hayama, Japan
| | - Teruo Kawada
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan.,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Laboratory of Molecular Function of Food, Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan; .,Research Unit for Physiological Chemistry, Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| |
Collapse
|
349
|
Cell-cycle arrest in mature adipocytes impairs BAT development but not WAT browning, and reduces adaptive thermogenesis in mice. Sci Rep 2017; 7:6648. [PMID: 28751675 PMCID: PMC5532220 DOI: 10.1038/s41598-017-07206-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 06/26/2017] [Indexed: 01/26/2023] Open
Abstract
We previously reported brown adipocytes can proliferate even after differentiation. To test the involvement of mature adipocyte proliferation in cell number control in fat tissue, we generated transgenic (Tg) mice over-expressing cell-cycle inhibitory protein p27 specifically in adipocytes, using the aP2 promoter. While there was no apparent difference in white adipose tissue (WAT) between wild-type (WT) and Tg mice, the amount of brown adipose tissue (BAT) was much smaller in Tg mice. Although BAT showed a normal cellular morphology, Tg mice had lower content of uncoupling protein 1 (UCP1) as a whole, and attenuated cold exposure- or β3-adrenergic receptor (AR) agonist-induced thermogenesis, with a decrease in the number of mature brown adipocytes expressing proliferation markers. An agonist for the β3-AR failed to increase the number of proliferating brown adipocytes, UCP1 content in BAT, and oxygen consumption in Tg mice, although the induction and the function of beige adipocytes in inguinal WAT from Tg mice were similar to WT mice. These results show that brown adipocyte proliferation significantly contributes to BAT development and adaptive thermogenesis in mice, but not to induction of beige adipocytes.
Collapse
|
350
|
Fat-enriched rather than high-fructose diets promote whitening of adipose tissue in a sex-dependent manner. J Nutr Biochem 2017; 49:22-29. [PMID: 28863366 DOI: 10.1016/j.jnutbio.2017.07.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 06/25/2017] [Accepted: 07/19/2017] [Indexed: 12/12/2022]
Abstract
Adipose tissue is a critical regulator of energy metabolism and an effector organ of excessive caloric intake. We studied the effects of high-fructose (HFruD), high-fat (HFD) and mixed high-sucrose and high-fat diet (HFHSD) on adipocyte morphology and biology and consecutive metabolic effects in male and female C57BL/6 mice. Forty male and 40 female mice were randomly assigned to one of four dietary groups and fed for 10 weeks ad libitum. After 10 weeks of feeding, mice were analyzed in regard to glucose metabolism, insulin sensitivity and alteration in adipocyte morphology and function. Weight gain and diminished insulin sensitivity in HFD- and HFHSD-fed mice were accompanied by increased adipocyte size and a shift in size distribution towards larger adipocytes in all mice. The latter effect was also found but less pronounced in HFruD-fed mice, while insulin sensitivity and body weight remained unaffected. In male mice, expansion of white adipocytes along with decreased uncoupling protein 1 (UCP-1) expression and alterations of mitochondrial biogenesis was found after HFD and HFHSD feeding, while in female mice, UCP-1 expression was also reduced in the HFruD dietary group. Diet-induced cellular alterations were less pronounced in female mice. Our data demonstrate that high-fat rather than high fructose consumption drives metabolically disadvantageous alterations of adipocyte differentiation involving whitening and insulin resistance in a sex-dependent manner with most deleterious effects seen upon administration of combined sucrose and fat-enriched diet in male mice.
Collapse
|