1
|
Li H, Li J, Song C, Yang H, Luo Q, Chen M. Brown adipose tissue: a potential target for aging interventions and healthy longevity. Biogerontology 2024; 25:1011-1024. [PMID: 39377866 DOI: 10.1007/s10522-024-10137-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/30/2024] [Indexed: 10/09/2024]
Abstract
Brown Adipose Tissue (BAT) is a type of fat tissue that can generate heat and plays an important role in regulating body temperature and energy metabolism. Enhancing BAT activity through medication, exercise and other means has become a potential effective method for treating metabolic disorders. Recently, there has been increasing evidence suggesting a link between BAT and aging. As humans age, the volume and activity of BAT decrease, which may contribute to the development of age-related diseases. Multiple organelles signaling pathways have been reported to be involved in the aging process associated with BAT. Therefore, we aimed to review the evidence related to the association between aging process and BAT decreasing, analyze the potential of BAT as a predictive marker for age-related diseases, and explore potential therapeutic strategies targeting BAT for aging interventions and healthy longevity.
Collapse
Affiliation(s)
- Hongde Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, PR China
| | - Junli Li
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China
| | - Chengxiang Song
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, PR China
| | - Haoran Yang
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, PR China
| | - Qiang Luo
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, PR China.
| | - Mao Chen
- Laboratory of Cardiac Structure and Function, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu, 610041, PR China.
- Department of Cardiology, West China Hospital, Sichuan University, #37 Guoxue Alley, Chengdu, 610041, PR China.
| |
Collapse
|
2
|
Fatriani R, Pratiwi FAK, Annisa A, Septaningsih DA, Aziz SA, Miladiyah I, Kusumastuti SA, Nasution MAF, Ramadhan D, Kusuma WA. Unveiling the anti-obesity potential of Kemuning (Murraya paniculata): A network pharmacology approach. PLoS One 2024; 19:e0305544. [PMID: 39208245 PMCID: PMC11361609 DOI: 10.1371/journal.pone.0305544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 05/30/2024] [Indexed: 09/04/2024] Open
Abstract
Obesity has become a global issue that affects the emergence of various chronic diseases such as diabetes mellitus, dysplasia, heart disorders, and cancer. In this study, an integration method was developed between the metabolite profile of the active compound of Murraya paniculata and the exploration of the targeting mechanism of adipose tissue using network pharmacology, molecular docking, molecular dynamics simulation, and in vitro tests. Network pharmacology results obtained with the skyline query technique using a block-nested loop (BNL) showed that histone acetyltransferase p300 (EP300), peroxisome proliferator-activated receptor gamma (PPARG), and peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PPARGC1A) are potential targets for treating obesity. Enrichment analysis of these three proteins revealed their association with obesity, thermogenesis, energy metabolism, adipocytokines, fat cell differentiation, and glucose homeostasis. Metabolite profiling of M. paniculata leaves revealed sixteen active compounds, ten of which were selected for molecular docking based on drug-likeness and ADME results. Molecular docking results between PPARG and EP300 with the ten active compounds showed a binding affinity value of ≤ -5.0 kcal/mol in all dockings, indicating strong binding. The stability of the protein-ligand complex resulting from docking was examined using molecular dynamics simulations, and we observed the best average root mean square deviation (RMSD) of 0.99 Å for PPARG with trans-3-indoleacrylic acid, which was lower than with the native ligand BRL (2.02 Å). Furthermore, the RMSD was 2.70 Å for EP300 and the native ligand 99E, and the lowest RMSD with the ligand (1R,9S)-5-[(E)-2-(4-Chlorophenyl)vinyl]-11-(5-pyrimidinylcarbonyl)-7,11-diazatricyclo[7.3.1.02,7]trideca-2,4-dien-6-one was 3.33 Å. The in vitro tests to validate the potential of M. paniculata in treating obesity showed that there was a significant decrease in PPARG and EP300 gene expressions in 3T3-L1 mature adipocytes treated with M. paniculata ethanolic extract starting at concentrations 62.5 μg/ml and 15.625 μg/ml, respectively. These results indicate that M. paniculata can potentially treat obesity by disrupting adipocyte maturation and influencing intracellular lipid metabolism.
Collapse
Affiliation(s)
- Rizka Fatriani
- Tropical Biopharmaca Research Center, IPB University, Bogor, Indonesia
| | | | - Annisa Annisa
- Department of Computer Science, Faculty of Mathematics and Natural Sciences, IPB University, Bogor, Indonesia
| | - Dewi Anggraini Septaningsih
- Department of Chemistry, Faculty of Military Mathematics and Natural Sciences, Republic of Indonesia Defense University, Bogor, Indonesia
| | - Sandra Arifin Aziz
- Department of Agronomy and Horticulture, Faculty of Agriculture, IPB University, Bogor, Indonesia
| | | | - Siska Andrina Kusumastuti
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | | | - Donny Ramadhan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Bogor, Indonesia
| | - Wisnu Ananta Kusuma
- Tropical Biopharmaca Research Center, IPB University, Bogor, Indonesia
- Department of Computer Science, Faculty of Mathematics and Natural Sciences, IPB University, Bogor, Indonesia
| |
Collapse
|
3
|
Schaftenaar FH, van Dam AD, de Bruin G, Depuydt MA, de Mol J, Amersfoort J, Douna H, Meijer M, Kröner MJ, van Santbrink PJ, Bernabé Kleijn MN, van Puijvelde GH, Florea BI, Slütter B, Foks AC, Bot I, Rensen PC, Kuiper J. Immunoproteasomal Inhibition With ONX-0914 Attenuates Atherosclerosis and Reduces White Adipose Tissue Mass and Metabolic Syndrome in Mice. Arterioscler Thromb Vasc Biol 2024; 44:1346-1364. [PMID: 38660806 PMCID: PMC11188635 DOI: 10.1161/atvbaha.123.319701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 04/10/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Atherosclerosis is the major underlying pathology of cardiovascular disease and is driven by dyslipidemia and inflammation. Inhibition of the immunoproteasome, a proteasome variant that is predominantly expressed by immune cells and plays an important role in antigen presentation, has been shown to have immunosuppressive effects. METHODS We assessed the effect of ONX-0914, an inhibitor of the immunoproteasomal catalytic subunits LMP7 (proteasome subunit β5i/large multifunctional peptidase 7) and LMP2 (proteasome subunit β1i/large multifunctional peptidase 2), on atherosclerosis and metabolism in LDLr-/- and APOE*3-Leiden.CETP mice. RESULTS ONX-0914 treatment significantly reduced atherosclerosis, reduced dendritic cell and macrophage levels and their activation, as well as the levels of antigen-experienced T cells during early plaque formation, and Th1 cells in advanced atherosclerosis in young and aged mice in various immune compartments. Additionally, ONX-0914 treatment led to a strong reduction in white adipose tissue mass and adipocyte progenitors, which coincided with neutrophil and macrophage accumulation in white adipose tissue. ONX-0914 reduced intestinal triglyceride uptake and gastric emptying, likely contributing to the reduction in white adipose tissue mass, as ONX-0914 did not increase energy expenditure or reduce total food intake. Concomitant with the reduction in white adipose tissue mass upon ONX-0914 treatment, we observed improvements in markers of metabolic syndrome, including lowered plasma triglyceride levels, insulin levels, and fasting blood glucose. CONCLUSIONS We propose that immunoproteasomal inhibition reduces 3 major causes underlying cardiovascular disease, dyslipidemia, metabolic syndrome, and inflammation and is a new target in drug development for atherosclerosis treatment.
Collapse
MESH Headings
- Animals
- Atherosclerosis/pathology
- Atherosclerosis/prevention & control
- Atherosclerosis/drug therapy
- Atherosclerosis/immunology
- Atherosclerosis/genetics
- Atherosclerosis/metabolism
- Metabolic Syndrome/drug therapy
- Metabolic Syndrome/immunology
- Disease Models, Animal
- Adipose Tissue, White/metabolism
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/pathology
- Receptors, LDL/genetics
- Receptors, LDL/deficiency
- Proteasome Endopeptidase Complex/metabolism
- Mice, Inbred C57BL
- Male
- Proteasome Inhibitors/pharmacology
- Apolipoprotein E3/genetics
- Apolipoprotein E3/metabolism
- Aortic Diseases/prevention & control
- Aortic Diseases/pathology
- Aortic Diseases/genetics
- Aortic Diseases/enzymology
- Aortic Diseases/immunology
- Aortic Diseases/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Macrophages/immunology
- Plaque, Atherosclerotic
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Mice, Knockout, ApoE
- Mice
- Energy Metabolism/drug effects
- Oligopeptides
Collapse
Affiliation(s)
- Frank H. Schaftenaar
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Andrea D. van Dam
- Division of Endocrinology, Department of Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (A.D.D., P.C.N.R.)
| | - Gerjan de Bruin
- Department of Chemical Biology, Leiden Institute of Chemistry, the Netherlands (G.d.B., B.I.F.)
| | - Marie A.C. Depuydt
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Jill de Mol
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Jacob Amersfoort
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Hidde Douna
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Menno Meijer
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Mara J. Kröner
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Peter J. van Santbrink
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Mireia N.A. Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Gijs H.M. van Puijvelde
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Bogdan I. Florea
- Department of Chemical Biology, Leiden Institute of Chemistry, the Netherlands (G.d.B., B.I.F.)
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Amanda C. Foks
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| | - Patrick C.N. Rensen
- Division of Endocrinology, Department of Medicine, Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, the Netherlands (A.D.D., P.C.N.R.)
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, the Netherlands (F.H.S., M.A.C.D., J.d.M., J.A., H.D., M.M., M.J.K., P.J.v.S., M.N.A.B.K., G.H.M.v.P., B.S., A.C.F., I.B., J.K.)
| |
Collapse
|
4
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Jeon MS, Zhang KK, Xie L, Layden BT, Ong SG, Jiang Y. The Notch-PDGFRβ axis suppresses brown adipocyte progenitor differentiation in early post-natal mice. Dev Cell 2024; 59:1233-1251.e5. [PMID: 38569546 DOI: 10.1016/j.devcel.2024.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 12/08/2023] [Accepted: 03/08/2024] [Indexed: 04/05/2024]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively explored. Here, through in vivo lineage tracing and mouse modeling, we observed that platelet-derived growth factor receptor beta (PDGFRβ)+ pericytes give rise to developmental brown adipocytes but not to those in adult homeostasis. By contrast, T-box 18 (TBX18)+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis by downregulating PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates high-fat, high-sucrose (HFHS)-induced glucose and metabolic impairment in mice during their development and juvenile phases. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health.
Collapse
Affiliation(s)
- Zuoxiao Shi
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Shaolei Xiong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Ruoci Hu
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zilai Wang
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jooman Park
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yanyu Qian
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Jaden Wang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Pratibha Bhalla
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Nipun Velupally
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Qing Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Minsun Stacey Jeon
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Ke Kurt Zhang
- Texas A&M Health Science Center, Institute of Biosciences and Technology, Houston, TX 77030, USA
| | - Linlin Xie
- Department of Nutrition, Texas A&M University, College Station, TX 77845, USA
| | - Brian T Layden
- Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Jesse Brown Medical VA Medical Center, Chicago, IL 60612, USA
| | - Sang-Ging Ong
- Department of Pharmacology and Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Cardiology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yuwei Jiang
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA; Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, USA; Division of Endocrinology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
5
|
Niclou A, Vesi L, Arorae M, Naseri NC, Savusa KF, Naseri T, Young J, Rivara AC, Ocobock C. Indication of mixed glucose and fatty acid use by inferred brown adipose tissue activity in Samoans. Am J Hum Biol 2024; 36:e23998. [PMID: 37823535 PMCID: PMC10939975 DOI: 10.1002/ajhb.23998] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVES Despite the growing rates of global obesity and the known positive associations between brown adipose tissue (BAT) and cardiovascular health, little is known about the metabolic effects of BAT activity in Samoans, a population at high risk of obesity and type II diabetes. Here we assessed the potential effects of inferred BAT activity on metabolic health markers in Samoan adults exposed to mild cold. METHODS Using point-of-care finger prick technology we measured fasting glucose, total cholesterol, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) levels before and after 30 min of cold exposure among 61 individuals (38 females, 23 males, ages 31-54) from 'Upolu Island, Samoa. Respiratory quotient was measured by indirect calorimetry to determine substrate metabolism at room temperature and cold exposure. RESULTS Fasting glucose levels decreased significantly (p < .001) after cold exposure while neither total cholesterol (p = .88), HDL (p = .312), nor LDL (p = .089) changed. Respiratory quotient decreased significantly (p = .009) between exposures, suggesting an increased preference for lipid metabolism as a response to cold. CONCLUSIONS The observed effects of inferred BAT activity on biomarkers suggest BAT activity utilizes both glucose and lipid-derived fatty acids as fuel for thermogenesis. Our work provides evidence for the beneficial metabolic effects of BAT and emphasizes the need for the population-specific development of metabolic treatments involving BAT to ensure the successful and equitable minimization of extreme consequences of obesity and metabolic health.
Collapse
Affiliation(s)
- Alexandra Niclou
- Pennington Biomedical Research Center, Baton Rouge, LA
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
| | - Lupesina Vesi
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | - Maria Arorae
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | | | | | | | - Jessica Young
- Center for Social Science Research, University of Notre Dame, Notre Dame, IN
| | - Anna C. Rivara
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
6
|
Niclou A, Vesi L, Arorae M, Naseri NC, Savusa KF, Naseri T, DeLany JP, McGarvey ST, Rivara AC, Ocobock C. When the cold gets under your skin: Evidence for brown adipose tissue activity in Samoan adults. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 183:e24848. [PMID: 37740598 PMCID: PMC10843446 DOI: 10.1002/ajpa.24848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/13/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023]
Abstract
OBJECTIVES Brown adipose tissue (BAT) is a heat-producing organ aiding nonshivering thermogenesis (NST) during cold stress. Due to its potential cold-adaptive role BAT has been predominantly studied in cold and temperate climate populations, but not among warm-climate adults. This work explores if BAT activity can be inferred in Samoans. MATERIALS AND METHODS We inferred BAT activity by comparing metabolic rate and surface heat dissipation using indirect calorimetry and thermal imaging between room temperature and cold exposure among Samoans (N = 61, females: n = 38) from 'Upolu Island, Samoa. BAT activity was inferred using ANOVA linear regression models with the variables measured at cold exposure as outcomes. T-tests were used to compare changes in surface temperature between room temperature and cold exposure. RESULTS Metabolic rate significantly increased after cooling. In both the supraclavicular area, a known BAT location, and the sternum, a non-BAT location, temperatures decreased significantly upon cold exposure. Differences in supraclavicular temperatures between room temperature and cold were significantly smaller than differences in sternum temperatures between exposures. These results suggest that BAT thermogenesis occurred in known BAT-locations and thus contributed to NST during cooling. CONCLUSIONS This study adds to our understanding of BAT activity across different populations and climates. Further study may illuminate whether the cold-adaptive properties of BAT may have played a role in the successful expansion of populations across the globe, including warm-climate groups.
Collapse
Affiliation(s)
- Alexandra Niclou
- Pennington Biomedical Research Center, Baton Rouge, LA
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
| | - Lupesina Vesi
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | - Maria Arorae
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | | | | | | | - James P. DeLany
- AdventHealth Orlando, Translational Research Institute, Orlando, FL
| | - Stephen T. McGarvey
- International Health Institute & Departments of Epidemiology and Anthropology, Brown University, Providence, RI
| | - Anna C. Rivara
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
7
|
Rao Y, Su R, Wu C, Yang G, Fu R, Wu J, Liang J, Liu J, Jiang Z, Xu C, Huang L. Marine fungus Aspergillus c1. sp metabolite activates the HSF1/PGC-1α axis, inducing a thermogenic program for treating obesity. Front Pharmacol 2024; 15:1320040. [PMID: 38333010 PMCID: PMC10851286 DOI: 10.3389/fphar.2024.1320040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Accepted: 01/15/2024] [Indexed: 02/10/2024] Open
Abstract
Background and aims: Obesity is one of the most prevalent diseases worldwide with less ideal approved agents in clinic. Activating the HSF1/PGC-1α axis in adipose tissues has been reported to induce thermogenesis in mice, which presents a promising therapeutic avenue for obesity treatment. The present study aimed to identified novel natural HSF1 activator and evaluated the therapeutic effects of the newly discovered compound on obesity-associated metabolic disorders and the molecular mechanisms of these effects. Methods: Our previous reported HSF1/PGC-1α activator screening system was used to identify novel natural HSF1 activator. The PGC-1α luciferase activity, immunoblot, protein nuclear-translocation, immunofluorescence, chromatin immunoprecipitation assays were used to evaluate the activity of compound HN-001 in activating HSF1. The experiments of mitochondrial number measurement, TG assay and imaging, cellular metabolic assay, gene assays, and CRISPR/Cas 9 were applied for investigating the metabolic effect of HN-001 in C3H10-T1/2 adipocytes. The in vivo anti-obesity efficacies and beneficial metabolic effects of HN-001 were evaluated by performing body and fat mass quantification, plasma chemical analysis, GTT, ITT, cold tolerance test, thermogenesis analysis. Results: HN-001 dose- and time-dependently activated HSF1 and induced HSF1 nuclear translocation, resulting in an enhancement in binding with the gene Pgc-1α. This improvement induced activation of adipose thermogenesis and enhancement of mitochondrial oxidation capacity, thus inhibiting adipocyte maturation. Deletion of HSF1 in adipocytes impaired mitochondrial oxidation and abolished the above beneficial metabolic effects of HN-001, including adipocyte browning induction, improvements in mitogenesis and oxidation capacity, and lipid-lowering ability. In mice, HN-001 treatment efficiently alleviated diet-induced obesity and metabolic disorders. These changes were associated with increased body temperature in mice and activation of the HSF1/PGC-1α axis in adipose tissues. UCP1 expression and mitochondrial biogenesis were increased in both white and brown adipose tissues of HN-001-treated mice. Conclusion: These data indicate that HN-001 may have therapeutic potential for obesity-related metabolic diseases by increasing the capacity of energy expenditure in adipose tissues through a mechanism involving the HSF1/PGC-1α axis, which shed new light on the development of novel anti-obesity agents derived from marine sources.
Collapse
Affiliation(s)
- Yong Rao
- *Correspondence: Yong Rao, ; Ling Huang,
| | | | | | | | | | | | | | | | | | | | - Ling Huang
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Hainan University, Haikou, China
| |
Collapse
|
8
|
Huang Z, Gu C, Zhang Z, Arianti R, Swaminathan A, Tran K, Battist A, Kristóf E, Ruan HB. Supraclavicular brown adipocytes originate from Tbx1+ myoprogenitors. PLoS Biol 2023; 21:e3002413. [PMID: 38048357 PMCID: PMC10721186 DOI: 10.1371/journal.pbio.3002413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 12/14/2023] [Accepted: 10/31/2023] [Indexed: 12/06/2023] Open
Abstract
Brown adipose tissue (BAT) dissipates energy as heat, contributing to temperature control, energy expenditure, and systemic homeostasis. In adult humans, BAT mainly exists in supraclavicular areas and its prevalence is associated with cardiometabolic health. However, the developmental origin of supraclavicular BAT remains unknown. Here, using genetic cell marking in mice, we demonstrate that supraclavicular brown adipocytes do not develop from the Pax3+/Myf5+ epaxial dermomyotome that gives rise to interscapular BAT (iBAT). Instead, the Tbx1+ lineage that specifies the pharyngeal mesoderm marks the majority of supraclavicular brown adipocytes. Tbx1Cre-mediated ablation of peroxisome proliferator-activated receptor gamma (PPARγ) or PR/SET Domain 16 (PRDM16), components of the transcriptional complex for brown fat determination, leads to supraclavicular BAT paucity or dysfunction, thus rendering mice more sensitive to cold exposure. Moreover, human deep neck BAT expresses higher levels of the TBX1 gene than subcutaneous neck white adipocytes. Taken together, our observations reveal location-specific developmental origins of BAT depots and call attention to Tbx1+ lineage cells when investigating human relevant supraclavicular BAT.
Collapse
Affiliation(s)
- Zan Huang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Chenxin Gu
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Zengdi Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Rini Arianti
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Aneesh Swaminathan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Kevin Tran
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Alex Battist
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Endre Kristóf
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| |
Collapse
|
9
|
Bielka W, Przezak A, Pawlik A. Follistatin and follistatin-like 3 in metabolic disorders. Prostaglandins Other Lipid Mediat 2023; 169:106785. [PMID: 37739334 DOI: 10.1016/j.prostaglandins.2023.106785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 09/02/2023] [Accepted: 09/16/2023] [Indexed: 09/24/2023]
Abstract
Follistatin (FST) is a glycoprotein which main role is antagonizing activity of transforming growth factor β superfamily members. Folistatin-related proteins such as follistatin-like 3 (FSTL3) also reveal these properties. The exact function of them has still not been established, but it can be bound to the pathogenesis of metabolic disorders. So far, there were performed a few studies about their role in type 2 diabetes, obesity or gestational diabetes and even less in type 1 diabetes. The outcomes are contradictory and do not allow to draw exact conclusions. In this article we summarize the available information about connections between follistatin, as well as follistatin-like 3, and metabolic disorders. We also emphasize the strong need of performing further research to explain their exact role, especially in the pathogenesis of diabetes and obesity.
Collapse
Affiliation(s)
- Weronika Bielka
- Department of Rheumatology and Internal Medicine, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Agnieszka Przezak
- Department of Rheumatology and Internal Medicine, Pomeranian Medical University in Szczecin, 71-252 Szczecin, Poland
| | - Andrzej Pawlik
- Department of Physiology, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland.
| |
Collapse
|
10
|
Shi Z, Xiong S, Hu R, Wang Z, Park J, Qian Y, Wang J, Bhalla P, Velupally N, Song Q, Song Z, Layden BT, Jiang Y. The Notch-Pdgfrβ axis suppresses brown adipocyte progenitor differentiation in early postnatal mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.24.541839. [PMID: 37293108 PMCID: PMC10245810 DOI: 10.1101/2023.05.24.541839] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively studied. Here through in vivo lineage tracing, we observed that PDGFRβ+ pericytes give rise to developmental brown adipocytes, but not to those in adult homeostasis. In contrast, TBX18+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRβ+ pericytes promotes brown adipogenesis through the downregulation of PDGFRβ. Furthermore, inhibition of Notch signaling in PDGFRβ+ pericytes mitigates HFHS (high-fat, high-sucrose) induced glucose and metabolic impairment in both developmental and adult stages. Collectively, these findings show that the Notch/PDGFRβ axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health. Highlights PDGFRβ+ pericytes act as an essential developmental brown APC.TBX18+ pericytes contribute to brown adipogenesis in a depot-specific manner.Inhibiting Notch-Pdgfrβ axis promotes brown APC adipogenesis.Enhanced postnatal brown adipogenesis improves metabolic health in adult stage.
Collapse
|
11
|
Yuan X, Lu H, Han M, Han K, Zhang Y, Liang P, Liu S, Cheng J. HCBP6-induced activation of brown adipose tissue and upregulated of BAT cytokines genes. J Therm Biol 2022; 109:103306. [DOI: 10.1016/j.jtherbio.2022.103306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/15/2022] [Accepted: 08/18/2022] [Indexed: 11/27/2022]
|
12
|
Lu PH, Chen YY, Tsai FM, Liao YL, Huang HF, Yu WH, Kuo CY. Combined Acupoints for the Treatment of Patients with Obesity: An Association Rule Analysis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:7252213. [PMID: 35341146 PMCID: PMC8947926 DOI: 10.1155/2022/7252213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 01/28/2022] [Accepted: 02/15/2022] [Indexed: 11/21/2022]
Abstract
Obesity is a prevalent metabolic disease that increases the risk of other diseases, such as hypertension, diabetes, hyperlipidemia, cardiovascular disease, and certain cancers. A meta-analysis of 11 randomized sham-controlled trials indicates that acupuncture had adjuvant benefits in improving simple obesity, and previous studies have reported that acupoint combinations were more useful than single-acupoint therapy. The Apriori algorithm, a data mining-based analysis that finds potential correlations in datasets, is broadly applied in medicine and business. This study, based on the Apriori algorithm-based association rule analysis, found the association rules of acupoints among 11 randomized controlled trials (RCTs). There were 23 acupoints extracted from 11 RCTs. We used Python to calculate the association between acupoints and disease. We found the top 10 frequency acupoints were Extra12, TF4, LI4, LI11, ST25, ST36, ST44, CO4, CO18, and CO1. We investigated the 1118 association rule and found that {LI4, ST36} ≥ {ST44}, {LI4, ST44} ≥ {ST36}, and {ST36, ST44} ≥ {LI4} were the most associated rules in the data. Acupoints, including LI4, ST36, and ST44, are the core acupoint combinations in the treatment of simple obesity.
Collapse
Affiliation(s)
- Ping-Hsun Lu
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yu-Yang Chen
- Department of Mathematics National Central University, Taoyuan, Taiwan
| | - Fu-Ming Tsai
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Yuan-Ling Liao
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Hui-Fen Huang
- Department of Chinese Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Post-Baccalaureate Chinese Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wei-Hsuan Yu
- Department of Mathematics National Central University, Taoyuan, Taiwan
| | - Chan-Yen Kuo
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| |
Collapse
|
13
|
Effect of Acupoint Catgut Embedding for Middle-Aged Obesity: A Multicentre, Randomised, Sham-Controlled Trial. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:4780019. [PMID: 35265146 PMCID: PMC8898815 DOI: 10.1155/2022/4780019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 01/10/2022] [Indexed: 11/17/2022]
Abstract
Objectives This study aimed to examine the efficacy and safety of acupoint catgut embedding (ACE) for obesity over a 16-week treatment period using sham stimulation as the control. Methods A multicenter, randomised, parallel, sham-controlled trial was conducted from February 10, 2017, to May 15, 2018. Men with waistlines ≥85 cm and women with ≥80 cm at three sites were randomised to receive eight sessions (over 16 weeks) of ACE (n = 108) or sham ACE (n = 108) with skin penetration at sham acupoints. The catgut was embedded once every two weeks using two alternating sets of acupoints. The follow-up lasted for an additional 24 weeks. The primary outcome was the percentage waistline reduction from baseline to week 16. Results We included 216 individuals in the intention-to-treat analysis. At 16 weeks, the rate of waistline reduction was 8.80% (95% confidence interval (CI), 7.93% to 9.66%) in the ACE group and 4.09% (95% CI, 3.18% to 5.00%) in the sham control group, with a between-group difference of 4.71% (95% CI, 3.47% to 5.95%; P < 0.0001). This difference persisted throughout the entire follow-up period (between-group difference after 24-week additional weeks, 4.94% (95% CI, 3.58% to 6.30%); P < 0.001). The subgroup analyses of waistline by sex (male/female) revealed treatment effects of 1.93 (95% CI, -0.37 to 4.23, P = 0.1) in the male group and 3.19 (95% CI, 1.99 to 4.39, P < 0.001) in the female group. The adverse event analysis suggested that ACE and laboratory tests confirmed the safety of ACE. Discussion. ACE for 16 weeks could decrease the waistline and weight and was safe for the treatment of obesity. Further research is needed to evaluate the long-term efficacy and sex differences. This trial is registered with NCT02936973.
Collapse
|
14
|
Lizcano F. Roles of estrogens, estrogen-like compounds, and endocrine disruptors in adipocytes. Front Endocrinol (Lausanne) 2022; 13:921504. [PMID: 36213285 PMCID: PMC9533025 DOI: 10.3389/fendo.2022.921504] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Women are subject to constitutional changes after menopause, which increases conditions and diseases prone to cardiovascular risks such as obesity and diabetes mellitus. Both estrogens and androgens influence the individual's metabolic mechanism, which controls the fat distribution and the hypothalamic organization of the regulatory centers of hunger and satiety. While androgens tend to accumulate fat in the splanchnic and the visceral region with an increase in cardiovascular risk, estrogens generate more subcutaneous and extremity distribution of adipose tissue. The absence of estrogen during menopause seems to be the main factor that gives rise to the greater predisposition of women to suffer cardiovascular alterations. However, the mechanisms by which estrogens regulate the energy condition of people are not recognized. Estrogens have several mechanisms of action, which mainly include the modification of specific receptors that belong to the steroid receptor superfamily. The alpha estrogen receptors (ERα) and the beta receptors (ERβ) have a fundamental role in the metabolic control of the individual, with a very characteristic corporal distribution that exerts an influence on the metabolism of lipids and glucose. Despite the significant amount of knowledge in this field, many of the regulatory mechanisms exerted by estrogens and ER continue to be clarified. This review will discuss the role of estrogens and their receptors on the central regulation of caloric expenditure and the influence they exert on the differentiation and function of adipocytes. Furthermore, chemical substances with a hormonal activity that cause endocrine disruption with affectation on estrogen receptors will be considered. Finally, the different medical therapies for the vasomotor manifestations of menopause and their role in reducing obesity, diabetes, and cardiovascular risk will be analyzed.
Collapse
|
15
|
Niclou A, Ocobock C. Weather permitting: Increased seasonal efficiency of nonshivering thermogenesis through brown adipose tissue activation in the winter. Am J Hum Biol 2021; 34:e23716. [PMID: 34942026 DOI: 10.1002/ajhb.23716] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES We investigated seasonal changes in brown adipose tissue (BAT) activation and metabolism in a temperate-climate Albany, NY population. METHODS Data were collected among 58 participants (21 males, 37 females, ages: 18-51) in the summer and 59 participants (23 males, 36 females, ages: 18-63) in the winter in Albany, New York. BAT activity was inferred by comparing metabolic rate, heat dissipation in the supraclavicular area, and respiratory quotient at room temperature and cold exposure. Seasonal variation in BAT was determined by comparing these measurements from summer and winter. RESULTS At mild cold exposure, heat dissipation of the supraclavicular area was significantly greater in the winter compared to summer (p < .001); however, no significant differences were found between seasons in metabolic rate measurements. This suggests BAT activation may be metabolically more efficient in the winter, due to prolonged lower seasonal temperatures relative to summer. Respiratory quotient significantly increased upon mild cold exposure in the winter compared to summer (p < .001). While carbohydrate utilization increased in the winter, fat remained the primary metabolic substrate for BAT activity across both seasons. CONCLUSION The seasonal variations in the effects of nonshivering thermogenesis on metabolic rate and substrate metabolism suggest a buffering of energy expenditure and an increased use of glucose as fuel by BAT as a result of acclimatization to cold in the winter. These findings point towards a potential role of BAT in human whole-body mediated glucose disposal and cold adaptation.
Collapse
Affiliation(s)
- Alexandra Niclou
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA
| | - Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, Indiana, USA
- Eck Institute for Global Health, Institute for Educational Initiatives, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
16
|
The New Role of AMP-Activated Protein Kinase in Regulating Fat Metabolism and Energy Expenditure in Adipose Tissue. Biomolecules 2021; 11:biom11121757. [PMID: 34944402 PMCID: PMC8698496 DOI: 10.3390/biom11121757] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/21/2021] [Accepted: 11/23/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is characterized by excessive accumulation of fat in the body, which is triggered by a body energy intake larger than body energy consumption. Due to complications such as cardiovascular diseases, type 2 diabetes (T2DM), obstructive pneumonia and arthritis, as well as high mortality, morbidity and economic cost, obesity has become a major health problem. The global prevalence of obesity, and its comorbidities is escalating at alarming rates, demanding the development of additional classes of therapeutics to reduce the burden of disease further. As a central energy sensor, the AMP-activated protein kinase (AMPK) has recently been elucidated to play a paramount role in fat synthesis and catabolism, especially in regulating the energy expenditure of brown/beige adipose tissue and the browning of white adipose tissue (WAT). This review discussed the role of AMPK in fat metabolism in adipose tissue, emphasizing its role in the energy expenditure of brown/beige adipose tissue and browning of WAT. A deeper understanding of the role of AMPK in regulating fat metabolism and energy expenditure can provide new insights into obesity research and treatment.
Collapse
|
17
|
Qiao A, Ma W, Deng J, Zhou J, Han C, Zhang E, Boriboun C, Xu S, Zhang C, Jie C, Kim JA, Habegger KM, Qiu H, Zhao TC, Zhang J, Qin G. Ablation of Sam68 in adult mice increases thermogenesis and energy expenditure. FASEB J 2021; 35:e21772. [PMID: 34252225 DOI: 10.1096/fj.202100021r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/27/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022]
Abstract
Genetic deletion of Src associated in mitosis of 68kDa (Sam68), a pleiotropic adaptor protein prevents high-fat diet-induced weight gain and insulin resistance. To clarify the role of Sam68 in energy metabolism in the adult stage, we generated an inducible Sam68 knockout mice. Knockout of Sam68 was induced at the age of 7-10 weeks, and then we examined the metabolic profiles of the mice. Sam68 knockout mice gained less body weight over time and at 34 or 36 weeks old, had smaller fat mass without changes in food intake and absorption efficiency. Deletion of Sam68 in mice elevated thermogenesis, increased energy expenditure, and attenuated core-temperature drop during acute cold exposure. Furthermore, we examined younger Sam68 knockout mice at 11 weeks old before their body weights deviate, and confirmed increased energy expenditure and thermogenic gene program. Thus, Sam68 is essential for the control of adipose thermogenesis and energy homeostasis in the adult.
Collapse
Affiliation(s)
- Aijun Qiao
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wenxia Ma
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianxin Deng
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Junlan Zhou
- Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Chaoshan Han
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eric Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chan Boriboun
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shiyue Xu
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chunxiang Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chunfa Jie
- Department of Biochemistry and Nutrition, Des Moines University College of Osteopathic Medicine, Des Moines, IA, USA
| | - Jeong-A Kim
- Department of Medicine-Endocrinology, Diabetes & Metabolism, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kirk M Habegger
- Department of Medicine-Endocrinology, Diabetes & Metabolism, Comprehensive Diabetes Center, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hongyu Qiu
- Center for Molecular and Translational Medicine, Institute of Biomedical Science, Georgia State University, Atlanta, GA, USA
| | - Ting C Zhao
- Department of Surgery, Boston University Medical School, Roger Williams Medical Center, Providence, RI, USA
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gangjian Qin
- Department of Biomedical Engineering, School of Medicine and School of Engineering, University of Alabama at Birmingham, Birmingham, AL, USA.,Feinberg Cardiovascular Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
18
|
Chen X, Xie J, Tan Q, Li H, Lu J, Zhang X. Genistein improves systemic metabolism and enhances cold resistance by promoting adipose tissue beiging. Biochem Biophys Res Commun 2021; 558:154-160. [PMID: 33915329 DOI: 10.1016/j.bbrc.2021.04.067] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/16/2021] [Indexed: 12/31/2022]
Abstract
Genistein, a naturally occurring phytoestrogen and a member of the large class of compounds known as isoflavones, exerts protective effects in several diseases. Recent studies indicate that genistein plays a critical role in controlling body weight, obesity-associated insulin resistance, and metabolic disorders, but its target organs in reversing obesity and related pathological conditions remain unclear. In this study, we showed that mice supplemented with 0.2% genistein in a high-fat diet for 12 weeks showed enhanced metabolic homeostasis, including reduced obesity, improved glucose uptake and insulin sensitivity, and alleviated hepatic steatosis. We also observed a beiging phenomenon in the white adipose tissue and reversal of brown adipose tissue whitening in these mice. These changes led to enhanced resistance to cold stress. Altogether, our data suggest that the improved metabolic profile in mice treated with genistein is likely a result of enhanced adipose tissue function.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, China; Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410010, China.
| | - Juanyu Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410010, China.
| | - Qingqing Tan
- Department of Biology, School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| | - Huan Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410010, China.
| | - Jun Lu
- Department of Pediatrics, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, 570208, China.
| | - Xingxing Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, 410010, China.
| |
Collapse
|
19
|
Atakan MM, Koşar ŞN, Güzel Y, Tin HT, Yan X. The Role of Exercise, Diet, and Cytokines in Preventing Obesity and Improving Adipose Tissue. Nutrients 2021; 13:nu13051459. [PMID: 33922998 PMCID: PMC8145589 DOI: 10.3390/nu13051459] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
The prevalence of obesity continues to rise worldwide despite evidence-based public health recommendations. The promise to adopt a healthy lifestyle is increasingly important for tackling this global epidemic. Calorie restriction or regular exercise or a combination of the two is accepted as an effective strategy in preventing or treating obesity. Furthermore, the benefits conferred by regular exercise to overcome obesity are attributed not only to reduced adiposity or reduced levels of circulating lipids but also to the proteins, peptides, enzymes, and metabolites that are released from contracting skeletal muscle or other organs. The secretion of these molecules called cytokines in response to exercise induces browning of white adipose tissue by increasing the expression of brown adipocyte-specific genes within the white adipose tissue, suggesting that exercise-induced cytokines may play a significant role in preventing obesity. In this review, we present research-based evidence supporting the effects of exercise and various diet interventions on preventing obesity and adipose tissue health. We also discuss the interplay between adipose tissue and the cytokines secreted from skeletal muscle and other organs that are known to affect adipose tissue and metabolism.
Collapse
Affiliation(s)
- Muhammed Mustafa Atakan
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Şükran Nazan Koşar
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Yasemin Güzel
- Division of Exercise Nutrition and Metabolism, Faculty of Sport Sciences, Hacettepe University, 06800 Ankara, Turkey; (M.M.A.); (Ş.N.K.); (Y.G.)
| | - Hiu Tung Tin
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
| | - Xu Yan
- Institute for Health and Sport (iHeS), Victoria University, P.O. Box 14428, Melbourne 8001, Australia;
- Sarcopenia Research Program, Australia Institute for Musculoskeletal Sciences (AIMSS), Melbourne 3021, Australia
- Correspondence: ; Tel.: +61-3-9919-4024; Fax: +61-3-9919-5615
| |
Collapse
|
20
|
Pervin S, Reddy ST, Singh R. Novel Roles of Follistatin/Myostatin in Transforming Growth Factor-β Signaling and Adipose Browning: Potential for Therapeutic Intervention in Obesity Related Metabolic Disorders. Front Endocrinol (Lausanne) 2021; 12:653179. [PMID: 33897620 PMCID: PMC8062757 DOI: 10.3389/fendo.2021.653179] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity is a global health problem and a major risk factor for several metabolic conditions including dyslipidemia, diabetes, insulin resistance and cardiovascular diseases. Obesity develops from chronic imbalance between energy intake and energy expenditure. Stimulation of cellular energy burning process has the potential to dissipate excess calories in the form of heat via the activation of uncoupling protein-1 (UCP1) in white and brown adipose tissues. Recent studies have shown that activation of transforming growth factor-β (TGF-β) signaling pathway significantly contributes to the development of obesity, and blockade or inhibition is reported to protect from obesity by promoting white adipose browning and increasing mitochondrial biogenesis. Identification of novel compounds that activate beige/brown adipose characteristics to burn surplus calories and reduce excess storage of fat are actively sought in the fight against obesity. In this review, we present recent developments in our understanding of key modulators of TGF-β signaling pathways including follistatin (FST) and myostatin (MST) in regulating adipose browning and brown adipose mass and activity. While MST is a key ligand for TGF-β family, FST can bind and regulate biological activity of several TGF-β superfamily members including activins, bone morphogenic proteins (BMP) and inhibins. Here, we review the literature supporting the critical roles for FST, MST and other proteins in modulating TGF-β signaling to influence beige and brown adipose characteristics. We further review the potential therapeutic utility of FST for the treatment of obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Shehla Pervin
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
| | - Srinivasa T. Reddy
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, United States
| | - Rajan Singh
- Department of Obstetrics and Gynecology, David Geffen School of Medicine at University of California Los Angeles (UCLA), Los Angeles, CA, United States
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, CA, United States
- Department of Endocrinology, Men’s Health: Aging and Metabolism, Brigham and Women’s Hospital, Boston, MA, United States
- *Correspondence: Rajan Singh,
| |
Collapse
|
21
|
Huang W, Chen X, Zhang Y, Wang L, Wang J, Zhang Y, Wei D, Zhou Z. Acupoint catgut embedding for obesity: A protocol of systematic review. Medicine (Baltimore) 2020; 99:e23728. [PMID: 33371124 PMCID: PMC7748162 DOI: 10.1097/md.0000000000023728] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 11/17/2020] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Obesity is a chronic metabolic disease in which patients are overweight due to the excessive accumulation of fat in the body. As a subtype of acupuncture, catgut embedding at acupoints has increased in clinical application for obesity. The aim of this study is to evaluate the effectiveness and safety of acupoint catgut embedding therapy for simple obesity. METHODS AND ANALYSIS Electronic searches of the Cochrane Library, PubMed, Springer Medline, EMBASE, Web of Science, China National Knowledge Infrastructure (CNKI), Wan-Fang Data (WANFANG), Chinese Biomedical Literature Database (CBM), and Chinese Scientific Journal Database (VIP databases) will be performed. The Chinese Clinical Trial Registry Center and the ClinicalTrials.gov registry will also be searched for ongoing trials. Databases will be searched from inception to August 2020.Randomized controlled clinical trials (RCTs) will be included if acupoint catgut embedding was evaluated as the sole treatment (diet or exercise therapy as the control group will be allowed) for simple obesity. The primary outcomes will consist of the improvement rate and reduction in body weight (BW). The secondary outcomes will include body mass index (BMI), waist circumference (WC), fat percentage (F %) and adverse effects. Two reviewers will undertake the study selection, data extraction and assessments of study quality. After screening the studies, the quality of the included studies will be assessed according to the quality criteria specified by the Cochrane Handbook for Systematic Reviews of Interventions (version 5.1.0). Meta-analysis will be performed by RevMan 5.3 software. RESULTS According to the data of improvement rate and reduction in BW, BMI, WC, and F %, this study will provide an evidence-based review of acupoint catgut embedding therapy for simply. CONCLUSIONS This systematic review will present the current evidence for acupoint catgut embedding therapy for obesity. ETHICS AND DISSEMINATION Ethical approval is not necessary since this protocol is only for systematic review and does not involve privacy data. The findings of this study will be disseminated electronically through a peer-review publication or presented at a relevant conference. TRIAL REGISTRATION NUMBER INPLASY2020110045.
Collapse
Affiliation(s)
- Wei Huang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Xia Chen
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yanji Zhang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province
| | - Lihua Wang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province
| | - Jiajie Wang
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yingrong Zhang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province
| | - Dan Wei
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Zhongyu Zhou
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
22
|
Chen X, Huang W, Wei D, Ding DG, Jiao Y, Pan HL, Jin YT, Zheng YW, Zhang YJ, Zhang YR, Liu YR, Zhou ZY. Clinical effect of catgut implantation at acupoints for the treatment of simple obesity: A multicentre randomized controlled trial. Medicine (Baltimore) 2020; 99:e23390. [PMID: 33235115 PMCID: PMC7710253 DOI: 10.1097/md.0000000000023390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 10/29/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Catgut implantation at acupoints (CIA) is a subtype of acupuncture that has been widely used to treat simple obesity, but evidence for its effectiveness remains scarce. The aim of this study is to evaluate the efficacy and safety of treating simple obesity with CIA. OBJECTIVE This clinical trial aims to evaluate the effectiveness and safety of CIA used for treatment of simple obesity. METHODS This is a multicentre, randomized, parallel, sham-controlled clinical trial. A total of 216 patients with simple obesity will be recruited. They will be randomly assigned in a 1:1 ratio to either the CIA group or the sham control group. All treatments will be given once every 2 weeks. The primary outcome measure is the rate of waistline reduction. Secondary outcome measures are the rates of reduction of body measurements, including weight, body mass index (BMI), hipline, waist-hip-ratio (WHR) and body fat percentage (BFP), the changes in scores on scales, including the Impact of Weight on Quality of Life Questionnaire (IWQOL-Lite), Short Form 36 (SF-36), the Hospital Anxiety and Depression Scale (HAD) and the Self-Esteem Scale (SES), Outcomes will be evaluated at baseline and at weeks 4, 8, 12, 16, 28, and 40, respectively. All adverse events that occur during this study will be recorded. If any participant withdraws from the trial, an intention-to-treat analysis (ITT) will be performed. CONCLUSION This is a randomized, sham-controlled trial of CIA treatment for simple obesity. The results of this trial will provide more evidence on whether CIA is efficacious and safe for treating obesity. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02936973. Registered on October 18, 2016.
Collapse
Affiliation(s)
- Xia Chen
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Wei Huang
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province, Wuhan, China
| | - Dan Wei
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - De-Guang Ding
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Yang Jiao
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Hong-Ling Pan
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Yi-Ting Jin
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province, Wuhan, China
| | - Yi-Wei Zheng
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Yan-Ji Zhang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province, Wuhan, China
| | - Ying-Rong Zhang
- Hubei University of Chinese Medicine/The Co-innovation Center for Preventive Treatment of Disease of Acupuncture-moxibustion in Hubei Province, Wuhan, China
| | - Yi-Ran Liu
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| | - Zhong-Yu Zhou
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine
- Hubei Province Academy of Traditional Chinese Medicine
| |
Collapse
|
23
|
Hao M, Guan Z, Gao Y, Xing J, Zhou X, Wang C, Xu J, Li W. Huang-Qi San ameliorates hyperlipidemia with obesity rats via activating brown adipocytes and converting white adipocytes into brown-like adipocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153292. [PMID: 32777487 DOI: 10.1016/j.phymed.2020.153292] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/05/2020] [Accepted: 07/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Brown adipose tissue (BAT) activation is a promising therapeutic target to treat hyperlipidemia with obesity. Huang-Qi San (HQS), an traditional Chinese medicine, can ameliorate hyperlipidemia with obesity, but its mechanism of action (MOA) is not understood. PURPOSE To articulate the MOA for HQS with animal models. METHODS The main chemical constituents of HQS were identified by high-performance liquid chromatography (HPLC) based assay. Hyperlipidemia with obesity rat models induced by high-fat diet were employed in the study. The levels of the fasting plasma glucose (FPG), triglyceride (TG), total cholesterol (TC), low-density lipoprotein-cholesterol (LDL-C) and high-density lipoprotein-cholesterol (HDL-C) were measured to evaluate the ability of HQS to ameliorate hyperlipidemia with obesity. Pathological analyses of organs were conducted with Oil Red O staining, hematoxylin-eosin (H&E) staining and transmission electron microscopy. The expression of mRNAs related to thermogenic genes, fatty acid oxidation-related genes and mitochondria biogenic genes were examined by quantitative real-time PCR. The protein expressions of uncoupling protein 1 (UCP1) were investigated by immunohistochemistry and western blot. Simultaneously, the protein expression of PR domain containing 16 (PRDM16), ATP synthase F1 subunit alpha (ATP5A) was detected by western blot. RESULTS HQS ameliorates metabolic disorder, lipid ectopic deposition, obesity and maintained glucose homeostasis in hyperlipidemia with obesity rats. HQS can significantly increase the number of mitochondria and reduced the size of the intracellular lipid droplets in BAT, and increase the expression of BAT activation-related genes (UCP1, PGC1α, PGC1β, Prdm16, CD137, TBX1, CPT1a, PPARα, Tfam, NRF1 and NRF2) in vivo. Furthermore, UCP1, PRDM16 and ATP5A proteins of BAT were increased. CONCLUSION HQS can activate BAT and browning of S-WAT (subcutaneous white adipose tissue) through activating the PRDM16/PGC1α/UCP1 pathway, augmenting mitochondrial biogenesis and fatty acid oxidation to increase thermogenesis and energy expenditure, resulting in a significant amelioration of hyperlipidemia with obesity. Therefore, HQS is an effective therapeutic medicine for the treatment of hyperlipidemia with obesity.
Collapse
Affiliation(s)
- Mengjiao Hao
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P.R. China; School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Zhuoji Guan
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510405, P.R. China
| | - Ying Gao
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Juling Xing
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Xinxin Zhou
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Chunyi Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China
| | - Jun Xu
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, P.R. China.
| | - Weimin Li
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou 510006, P.R. China.
| |
Collapse
|
24
|
Ruan HB. Developmental and functional heterogeneity of thermogenic adipose tissue. J Mol Cell Biol 2020; 12:775-784. [PMID: 32569352 PMCID: PMC7816678 DOI: 10.1093/jmcb/mjaa029] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/11/2020] [Accepted: 06/12/2020] [Indexed: 12/18/2022] Open
Abstract
The obesity epidemic continues to rise as a global health challenge. Thermogenic brown and beige adipocytes dissipate chemical energy as heat, providing an opportunity for developing new therapeutics for obesity and related metabolic diseases. Anatomically, brown adipose tissue is distributed as discrete depots, while beige adipocytes exist within certain depots of white adipose tissue. Developmentally, brown and beige adipocytes arise from multiple embryonic progenitor populations that are distinct and overlapping. Functionally, they respond to a plethora of stimuli to engage uncoupling protein 1-dependent and independent thermogenic programs, thus improving systemic glucose homeostasis, lipid metabolism, and the clearance of branched-chain amino acids. In this review, we highlight recent advances in our understanding of the molecular and cellular mechanisms that contribute to the developmental and functional heterogeneity of thermogenic adipose tissue.
Collapse
Affiliation(s)
- Hai-Bin Ruan
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
25
|
Ahmad B, Serpell CJ, Fong IL, Wong EH. Molecular Mechanisms of Adipogenesis: The Anti-adipogenic Role of AMP-Activated Protein Kinase. Front Mol Biosci 2020; 7:76. [PMID: 32457917 PMCID: PMC7226927 DOI: 10.3389/fmolb.2020.00076] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/03/2020] [Indexed: 12/24/2022] Open
Abstract
Obesity is now a widespread disorder, and its prevalence has become a critical concern worldwide, due to its association with common co-morbidities like cancer, cardiovascular diseases and diabetes. Adipose tissue is an endocrine organ and therefore plays a critical role in the survival of an individual, but its dysfunction or excess is directly linked to obesity. The journey from multipotent mesenchymal stem cells to the formation of mature adipocytes is a well-orchestrated program which requires the expression of several genes, their transcriptional factors, and signaling intermediates from numerous pathways. Understanding all the intricacies of adipogenesis is vital if we are to counter the current epidemic of obesity because the limited understanding of these intricacies is the main barrier to the development of potent therapeutic strategies against obesity. In particular, AMP-Activated Protein Kinase (AMPK) plays a crucial role in regulating adipogenesis – it is arguably the central cellular energy regulation protein of the body. Since AMPK promotes the development of brown adipose tissue over that of white adipose tissue, special attention has been given to its role in adipose tissue development in recent years. In this review, we describe the molecular mechanisms involved in adipogenesis, the role of signaling pathways and the substantial role of activated AMPK in the inhibition of adiposity, concluding with observations which will support the development of novel chemotherapies against obesity epidemics.
Collapse
Affiliation(s)
- Bilal Ahmad
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | | | - Isabel Lim Fong
- Department of Paraclinical Sciences, Faculty of Medicine and Health Sciences, Universiti Malaysia Sarawak, Kota Samarahan, Malaysia
| | - Eng Hwa Wong
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| |
Collapse
|
26
|
|
27
|
Li X, Wang G, Liu J, Ding G. Increased UCP1 expression in the perirenal adipose tissue of patients with renal cell carcinoma. Oncol Rep 2019; 42:1972-1980. [PMID: 31545449 PMCID: PMC6775817 DOI: 10.3892/or.2019.7306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 08/07/2019] [Indexed: 11/22/2022] Open
Abstract
Perirenal adipose tissue (PAT) has been implicated in renal cell carcinoma (RCC). The expression of uncoupling protein 1 (UCP1) is higher in PAT compared with that in back subcutaneous adipose tissue (bSAT). The aim of the present study was to determine UCP1 expression in different parts of PAT and to analyze the correlation between UCP1 expression in PAT and RCC. PAT from the upper and lower renal poles and bSAT samples were collected from 50 patients with RCC (RCC group) and 54 patients with renal cysts (control group) who had undergone renal surgery. Both UCP1 mRNA and protein levels were found to be significantly higher and adipocytes appeared to be smaller in the PAT of the RCC group. Furthermore, the RCC group had more multilocular UCP1-positive adipocytes. UCP1 staining in the PAT was significantly stronger in the RCC group, but there was no significant difference in UCP1 staining in the bSAT between the two groups. Furthermore, Fuhrman grade and T stage were higher in the high UCP1 expression group of RCC patients. In conclusion, high UCP1 expression in the PAT may serve as an indicator of poor prognosis in RCC.
Collapse
Affiliation(s)
- Xueqin Li
- Department of Gerontology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Gongcheng Wang
- Department of Urology, The Affiliated Huai'an No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Juan Liu
- Department of Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| | - Guoxian Ding
- Department of Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
28
|
Park J, Kim HL, Jung Y, Ahn KS, Kwak HJ, Um JY. Bitter Orange (Citrus aurantium Linné) Improves Obesity by Regulating Adipogenesis and Thermogenesis through AMPK Activation. Nutrients 2019; 11:nu11091988. [PMID: 31443565 PMCID: PMC6770725 DOI: 10.3390/nu11091988] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/09/2019] [Accepted: 08/18/2019] [Indexed: 12/19/2022] Open
Abstract
Obesity is a global health threat. Herein, we evaluated the underlying mechanism of anti-obese features of bitter orange (Citrus aurantium Linné, CA). Eight-week-administration of CA in high fat diet-induced obese C57BL/6 mice resulted in a significant decrease of body weight, adipose tissue weight and serum cholesterol. In further in vitro studies, we observed decreased lipid droplets in CA-treated 3T3-L1 adipocytes. Suppressed peroxisome proliferator-activated receptor gamma (PPARγ) and CCAAT/enhancer binding protein alpha indicated CA-inhibited adipogenesis. Moreover, CA-treated primary cultured brown adipocytes displayed increased differentiation associated with elevation of thermogenic factors including uncoupling protein 1 and PPARγ coactivator 1 alpha as well. The effects of CA in both adipocytes were abolished in AMP-activated protein kinase alpha (AMPKα)-suppressed environments, suggesting the anti-adipogenic and pro-thermogenic actions of CA were dependent on AMPKα pathway. In conclusion, our results suggest CA as a potential anti-obese agent which regulates adipogenesis and thermogenesis via AMPKα.
Collapse
Affiliation(s)
- Jinbong Park
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Basic Research Laboratory for Comorbidity Regulation, Comorbidity Research Institute, Kyung Hee University, Seoul 02447, Korea
| | - Hye-Lin Kim
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yunu Jung
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Basic Research Laboratory for Comorbidity Regulation, Comorbidity Research Institute, Kyung Hee University, Seoul 02447, Korea
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea
- Basic Research Laboratory for Comorbidity Regulation, Comorbidity Research Institute, Kyung Hee University, Seoul 02447, Korea
| | - Hyun Jeong Kwak
- Life science major, Division of Bio-convergence, College of convergence and integrate science, Kyonggi University, Suwon 16227, Korea.
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, Seoul 02447, Korea.
- Basic Research Laboratory for Comorbidity Regulation, Comorbidity Research Institute, Kyung Hee University, Seoul 02447, Korea.
| |
Collapse
|
29
|
Potential exerkines for physical exercise-elicited pro-cognitive effects: Insight from clinical and animal research. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2019; 147:361-395. [PMID: 31607361 DOI: 10.1016/bs.irn.2019.06.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
A sedentary lifestyle is now known as a critical risk factor for accelerated aging-related neurodegenerative disorders. In contract, having regular physical exercise has opposite effects. Clinical findings have suggested that physical exercise can promote brain plasticity, particularly the hippocampus and the prefrontal cortex, that are important for learning and memory and mood regulations. However, the underlying mechanisms are still unclear. Animal studies reveal that the effects of physical exercise on promoting neuroplasticity could be mediated by different exerkines derived from the peripheral system and the brain itself. This book chapter summarizes the recent evidence from clinical and pre-clinical studies showing the emerging mediators for exercise-promoted brain health, including myokines secreted from skeletal muscles, adipokines from adipose tissues, and other factors secreted from the bone and liver.
Collapse
|
30
|
Palmer BF, Clegg DJ. Strategies to Counter Weight Loss-Induced Reductions in Metabolic Rate. Curr Sports Med Rep 2019; 18:258-265. [DOI: 10.1249/jsr.0000000000000610] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
31
|
Ganeff IMM, Bos MM, van Heemst D, Noordam R. BMI-associated gene variants in FTO and cardiometabolic and brain disease: obesity or pleiotropy? Physiol Genomics 2019; 51:311-322. [PMID: 31199196 DOI: 10.1152/physiolgenomics.00040.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Obesity is a causal risk factor for the development of age-related disease conditions, which includes Type 2 diabetes mellitus, cardiovascular disease, and dementia. In genome-wide association studies, genetic variation in FTO is strongly associated with obesity and has been described across different ethnic backgrounds and life stages. To date, much work has been devoted on determining the biological mechanisms via which FTO affects body weight regulation and ultimately contributes to age-related cardiometabolic and brain disease. The main hypotheses of the involved biological mechanisms include the involvement of FTO in habitual food intake and energy expenditure. In this narrative review, our overall aim is to provide an overview on how FTO gene variants could increase the risk of developing age-related disease conditions. Specifically, we will discuss the state of the literature based on the different hypotheses how FTO regulates body weight and ultimately contributes to cardiometabolic disease and brain disease.
Collapse
Affiliation(s)
- Ingeborg M M Ganeff
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Maxime M Bos
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Diana van Heemst
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| | - Raymond Noordam
- Department of Internal Medicine, Section Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
32
|
Wang LH, Huang W, Wei D, Ding DG, Liu YR, Wang JJ, Zhou ZY. Mechanisms of Acupuncture Therapy for Simple Obesity: An Evidence-Based Review of Clinical and Animal Studies on Simple Obesity. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2019; 2019:5796381. [PMID: 30854010 PMCID: PMC6378065 DOI: 10.1155/2019/5796381] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/25/2018] [Indexed: 12/15/2022]
Abstract
Simple obesity is a worldwide epidemic associated with rapidly growing morbidity and mortality which imposes an enormous burden on individual and public health. As a part of Traditional Chinese Medicine (TCM), acupuncture has shown the positive efficacy in the management of simple obesity. In this article, we comprehensively review the clinical and animal studies that demonstrated the potential mechanisms of acupuncture treatment for simple obesity. Clinical studies suggested that acupuncture regulates endocrine system, promotes digestion, attenuates oxidative stress, and modulates relevant molecules of metabolism in patients of simple obesity. Evidence from laboratory indicated that acupuncture regulates lipid metabolism, modulates inflammatory responses, and promotes white adipose tissue browning. Acupuncture also suppresses appetite through regulating appetite regulatory hormones and the downstream signaling pathway. The evidence from clinical and animal studies indicates that acupuncture induces multifaceted regulation through complex mechanisms and moreover a single factor may not be enough to explain the beneficial effects against simple obesity.
Collapse
Affiliation(s)
- Li-Hua Wang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
| | - Wei Huang
- College of Acupuncture and Orthopedics, Hubei University of Chinese Medicine/Hubei Provincial Collaborative Innovation Center of Preventive Treatment by Acupuncture and Moxibustion, Wuhan, China
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Dan Wei
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - De-Guang Ding
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Yi-Ran Liu
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Jia-Jie Wang
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Zhong-Yu Zhou
- Department of Acupuncture, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
33
|
Zoico E, Rubele S, De Caro A, Nori N, Mazzali G, Fantin F, Rossi A, Zamboni M. Brown and Beige Adipose Tissue and Aging. Front Endocrinol (Lausanne) 2019; 10:368. [PMID: 31281288 PMCID: PMC6595248 DOI: 10.3389/fendo.2019.00368] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 05/23/2019] [Indexed: 01/24/2023] Open
Abstract
Across aging, adipose tissue (AT) changes its quantity and distribution: AT becomes dysfunctional with an increase in production of inflammatory peptides, a decline of those with anti-inflammatory activity and infiltration of macrophages. Adipose organ dysfunction may lead to age-related metabolic alterations. Aging is characterized by an increase in adiposity and a decline in brown adipose tissue (BAT) depots and activity, and UCP1 expression. There are many possible links to age-associated involution of BAT, including the loss of mitochondrial function, impairment of the sympathetic nervous system, age-induced alteration of brown adipogenic stem/progenitor cell function and changes in endocrine signals. Aging is also associated with a reduction in beige adipocyte formation. Beige adipocytes are known to differentiate from a sub-population of progenitors resident in white adipose tissue (WAT); a defective ability of progenitor cells to proliferate and differentiate has been hypothesized with aging. The loss of beige adipocytes with age may be caused by changes in trophic factors in the adipose tissue microenvironment, which regulate progenitor cell proliferation and differentiation. This review focuses on possible mechanisms involved in the reduction of BAT and beige activity with aging, along with possible targets for age-related metabolic disease therapy.
Collapse
Affiliation(s)
- Elena Zoico
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
- *Correspondence: Elena Zoico
| | - Sofia Rubele
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Annamaria De Caro
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Nicole Nori
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Gloria Mazzali
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Francesco Fantin
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Andrea Rossi
- Division of Geriatric Medicine, Department of Medicine, University of Verona, Verona, Italy
| | - Mauro Zamboni
- Division of Geriatric Medicine, Department of Surgery, Dentistry, Pediatric and Gynecology, University of Verona, Verona, Italy
| |
Collapse
|
34
|
Wollina U, Goldman A, Abdel-Naser MB, Philipp-Dormston WG. Adipose tissue, fillers, and skin tightening. Dermatol Ther 2018; 32:e12626. [PMID: 30175548 DOI: 10.1111/dth.12626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/24/2018] [Indexed: 12/15/2022]
Abstract
The role of adipose tissue has long been underestimated in esthetic dermatology. With the development of liposculpture and lipolysis, subcutaneous adipose tissue has gained an increasing interest. Harvested tissue has been used for lipofilling. In recent years, a better understanding of adipocyte physiology and its role in aging opened a new road for targeted treatments. Subcutaneous adipose tissue is no longer an innocent bystander in the combat of aging and the correction in esthetics. Adipose tissue is of importance for metabolic function and thermoregulation. Adipose tissue is involved in inflammation. Adipose tissue is heterogeneous in sense of function, color and size of adipocytes. The tissue is an important source of somatic stem cells.
Collapse
Affiliation(s)
- Uwe Wollina
- Department of Dermatology and Allergology, Academic Teaching Hospital Dresden, Dresden, Germany
| | - Alberto Goldman
- Department of Plastic Surgery, Clinica Goldman and Hospital Moinhos de Vento, Porto Alegre, Brazil
| | - Mohamed B Abdel-Naser
- Department of Dermatology and Venereology, Ain Shams University Hospital, Cairo, Egypt
| | | |
Collapse
|
35
|
Şahin M, Canpolat AG, Çorapçioğlu D, Canpolat U, Emral R, Uysal AR. Association between circulating irisin levels and epicardial fat in patients with treatment-naïve overt hyperthyroidism. Biomarkers 2018; 23:742-747. [PMID: 29862847 DOI: 10.1080/1354750x.2018.1485056] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND Hyperthyroidism is associated with increased metabolic activity and thermogenesis. Irisin is a key molecule in thermogenesis and energy expenditure via adipose tissue browning. Epicardial fat was previously defined as brown-like fat. Thus, here we aimed to evaluate the association between serum irisin level and epicardial fat thickness (EFT) in patients with hyperthyroidism. METHODS A total of 25 hyperthyroid patients and 24 age-, sex- and BMI-matched healthy controls were enrolled. Serum irisin levels, thyroid hormone levels, and body compositions were compared. EFT was measured via transthoracic echocardiography. RESULTS Serum irisin level and EFT were significantly higher in the hyperthyroid group (p < 0.001 and p = 0.001, respectively). The distributions of fat-free mass, muscle mass and fat mass were similar between the study groups. Serum irisin level was negatively correlated with TSH (p < 0.001) and positively correlated with fT3 (p < 0.001), fT4 (p < 0.001) and TSH receptor antibody (p = 0.002) levels and EFT (p = 0.001). In multivariate linear regression analysis, TSH (β = -0.475, p < 0.001) and EFT (β = 0.290, p = 0.023) levels were significantly associated with serum irisin levels. CONCLUSIONS An increased serum irisin level associated with EFT might contribute to metabolic derangement in hyperthyroidism. Further studies are needed to elucidate whether irisin levels and EFT are affected by hyperthyroidism or vice versa.
Collapse
Affiliation(s)
- Mustafa Şahin
- a Department of Endocrinology and Metabolism , Ankara University Faculty of Medicine , Ankara , Turkey
| | - Asena Gökçay Canpolat
- a Department of Endocrinology and Metabolism , Ankara University Faculty of Medicine , Ankara , Turkey
| | - Demet Çorapçioğlu
- a Department of Endocrinology and Metabolism , Ankara University Faculty of Medicine , Ankara , Turkey
| | - Uğur Canpolat
- b Department of Cardiology , Hacettepe University Faculty of Medicine , Ankara , Turkey
| | - Rıfat Emral
- a Department of Endocrinology and Metabolism , Ankara University Faculty of Medicine , Ankara , Turkey
| | - Ali Rıza Uysal
- a Department of Endocrinology and Metabolism , Ankara University Faculty of Medicine , Ankara , Turkey
| |
Collapse
|
36
|
Marlatt KL, Chen KY, Ravussin E. Is activation of human brown adipose tissue a viable target for weight management? Am J Physiol Regul Integr Comp Physiol 2018; 315:R479-R483. [PMID: 29741929 DOI: 10.1152/ajpregu.00443.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
To date, human studies show that brown adipose tissue (BAT) contributes a small yet highly variable amount to overall energy expenditure. No studies have shown a decrease in body weight with cold-induced BAT activation, and existing pharmacological studies suggest that BAT activation via the sympathetic nervous system may result in increased heart rate and systolic blood pressure. Furthermore, even though the amount and/or activity of BAT have been shown to vary with seasons, such variation does not seem to be translated into weight changes. Collectively, these findings do not support the use of BAT activation for weight loss in humans; however, the potential role of BAT in counteracting the metabolic adaptation observed with weight loss is suggested. Although the role of BAT in weight control is currently unsubstantiated, BAT may play a role in improving insulin sensitivity in humans.
Collapse
Affiliation(s)
- Kara L Marlatt
- Pennington Biomedical Research Center , Baton Rouge, Louisiana
| | - Kong Y Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health , Bethesda, Maryland
| | - Eric Ravussin
- Pennington Biomedical Research Center , Baton Rouge, Louisiana
| |
Collapse
|
37
|
Panagia M, Chen HH, Croteau D, Iris Chen YC, Ran C, Luptak I, Josephson L, Colucci WS, Sosnovik DE. Multiplexed Optical Imaging of Energy Substrates Reveals That Left Ventricular Hypertrophy Is Associated With Brown Adipose Tissue Activation. Circ Cardiovasc Imaging 2018; 11:e007007. [PMID: 29555834 PMCID: PMC5908227 DOI: 10.1161/circimaging.117.007007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 01/18/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND Substrate utilization in tissues with high energetic requirements could play an important role in cardiometabolic disease. Current techniques to assess energetics are limited by high cost, low throughput, and the inability to resolve multiple readouts simultaneously. Consequently, we aimed to develop a multiplexed optical imaging platform to simultaneously assess energetics in multiple organs in a high throughput fashion. METHODS AND RESULTS The detection of 18F-Fluordeoxyglucose uptake via Cerenkov luminescence and free fatty acid uptake with a fluorescent C16 free fatty acid was tested. Simultaneous uptake of these agents was measured in the myocardium, brown/white adipose tissue, and skeletal muscle in mice with/without thoracic aortic banding. Within 5 weeks of thoracic aortic banding, mice developed left ventricular hypertrophy and brown adipose tissue activation with upregulation of β3AR (β3 adrenergic receptors) and increased natriuretic peptide receptor ratio. Imaging of brown adipose tissue 15 weeks post thoracic aortic banding revealed an increase in glucose (P<0.01) and free fatty acid (P<0.001) uptake versus controls and an increase in uncoupling protein-1 (P<0.01). Similar but less robust changes were seen in skeletal muscle, while substrate uptake in white adipose tissue remained unchanged. Myocardial glucose uptake was increased post-thoracic aortic banding but free fatty acid uptake trended to decrease. CONCLUSIONS A multiplexed optical imaging technique is presented that allows substrate uptake to be simultaneously quantified in multiple tissues in a high throughput manner. The activation of brown adipose tissue occurs early in the onset of left ventricular hypertrophy, which produces tissue-specific changes in substrate uptake that may play a role in the systemic response to cardiac pressure overload.
Collapse
Affiliation(s)
- Marcello Panagia
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Howard H Chen
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Dominique Croteau
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Yin-Ching Iris Chen
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Chongzhao Ran
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Ivan Luptak
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Lee Josephson
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - Wilson S Colucci
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA
| | - David E Sosnovik
- From the Cardiovascular Medicine Section, Department of Medicine, Boston University Medical Center, MA (M.P., D.C., I.L., W.S.C.); Cardiovascular Research Center (M.P., H.H.C., D.E.S.) and Martinos Center for Biomedical Imaging, Department of Radiology (M.P., H.H.C., Y.-C.I.C., C.R., L.J., D.E.S.), Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
38
|
Wu L, Zhang L, Li B, Jiang H, Duan Y, Xie Z, Shuai L, Li J, Li J. AMP-Activated Protein Kinase (AMPK) Regulates Energy Metabolism through Modulating Thermogenesis in Adipose Tissue. Front Physiol 2018. [PMID: 29515462 PMCID: PMC5826329 DOI: 10.3389/fphys.2018.00122] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Obesity occurs when excess energy accumulates in white adipose tissue (WAT), whereas brown adipose tissue (BAT), which is specialized in dissipating energy through thermogenesis, potently counteracts obesity. White adipocytes can be converted to thermogenic "brown-like" cells (beige cells; WAT browning) under various stimuli, such as cold exposure. AMP-activated protein kinase (AMPK) is a crucial energy sensor that regulates energy metabolism in multiple tissues. However, the role of AMPK in adipose tissue function, especially in the WAT browning process, is not fully understood. To illuminate the effect of adipocyte AMPK on energy metabolism, we generated Adiponectin-Cre-driven adipose tissue-specific AMPK α1/α2 KO mice (AKO). These AKO mice were cold intolerant and their inguinal WAT displayed impaired mitochondrial integrity and biogenesis, and reduced expression of thermogenic markers upon cold exposure. High-fat-diet (HFD)-fed AKO mice exhibited increased adiposity and exacerbated hepatic steatosis and fibrosis and impaired glucose tolerance and insulin sensitivity. Meanwhile, energy expenditure and oxygen consumption were markedly decreased in the AKO mice both in basal conditions and after stimulation with a β3-adrenergic receptor agonist, CL 316,243. In contrast, we found that in HFD-fed obese mouse model, chronic AMPK activation by A-769662 protected against obesity and related metabolic dysfunction. A-769662 alleviated HFD-induced glucose intolerance and reduced body weight gain and WAT expansion. Notably, A-769662 increased energy expenditure and cold tolerance in HFD-fed mice. A-769662 treatment also induced the browning process in the inguinal fat depot of HFD-fed mice. Likewise, A-769662 enhanced thermogenesis in differentiated inguinal stromal vascular fraction (SVF) cells via AMPK signaling pathway. In summary, a lack of adipocyte AMPKα induced thermogenic impairment and obesity in response to cold and nutrient-overload, respectively, whereas chronic AMPK activation by A-769662 promoted WAT browning in inguinal WAT and protected against HFD-induced obesity and related metabolic dysfunction. These findings reveal a vital role for adipocyte AMPK in regulating the browning process in inguinal WAT and in maintaining energy homeostasis, which suggests that the targeted activation of adipocyte AMPK may be a promising strategy for anti-obesity therapy.
Collapse
Affiliation(s)
- Lingyan Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lina Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Bohan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Haowen Jiang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China
| | - Yanan Duan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, East China Normal University, Shanghai, China
| | - Zhifu Xie
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Lin Shuai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jingya Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
39
|
Abstract
PURPOSE OF REVIEW New treatment approaches to weight loss and weight loss maintenance in humans are critical. Given its potential role in stimulating energy expenditure, brown adipose tissue (BAT) activation has become a trending topic as an anti-obesity treatment. RECENT FINDINGS Most studies on BAT stimulation have been conducted in rodents and used cold stimulation. To date, few human trials exist that tested the effect of cold exposure on BAT. Those studies show that BAT contributes a small amount to overall energy metabolism which is unlikely to cause weight loss. Nonetheless, improvements in glucose metabolism have been demonstrated in humans. While new pharmacological approaches demonstrate some contribution of BAT to overall energy expenditure, the potential cardiovascular risk (increased heart rate and blood pressure to sustain the extra energy expenditure) may preclude their use. There is no convincing evidence yet to indicate that BAT may be a viable pharmaceutical target for body weight loss or even weight loss maintenance. More research is needed to confirm the relevance of BAT and beige tissue to whole-body energy metabolism in humans.
Collapse
Affiliation(s)
- Kara L Marlatt
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
40
|
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: 7.7] [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
|
41
|
Frank AP, Palmer BF, Clegg DJ. Do estrogens enhance activation of brown and beiging of adipose tissues? Physiol Behav 2017; 187:24-31. [PMID: 28988965 DOI: 10.1016/j.physbeh.2017.09.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/30/2017] [Accepted: 09/30/2017] [Indexed: 01/05/2023]
Abstract
Obesity and its associated co-morbidities are worldwide public health concerns. Obesity is characterized by excessive adipose tissue accumulation; however, it is important to recognize that human and rodent adipose tissues are made up of several distinct adipose tissue sub-types. White adipose tissue (WAT) is considered the prototypical fat cell, due to its capacity and capability to store large amounts of lipid. In contrast, brown adipose tissue (BAT) oxidizes substrates to generate heat. BAT contains more mitochondria than WAT and express uncoupling protein-1 (UCP1), which mediates BAT thermogenesis. A third sub-type of adipose tissue, Brown-in-white (BRITE)/beige adipocytes arise from WAT upon adrenergic stimulation and resembles BAT functionally. The energy burning feature of BAT/beige cells, combined with evidence of an inverse-correlation between BAT/beige adipose tissue and obesity have given rise to the hypothesis that obesity may be linked to BAT/beige 'malfunction'. Females have more BAT and perhaps an enhanced capacity to beige their adipose tissue when compared to males. Multiple signal pathways are capable of activating BAT thermogenesis and beiging of WAT; here, we discuss the potential role of estrogens in enhancing and mediating these factors to enhance adipose tissue thermogenesis.
Collapse
Affiliation(s)
- Aaron P Frank
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Biff F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Deborah J Clegg
- Biomedical Research Division, Diabetes and Obesity Research Institute, Department of Biomedical Science, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|
42
|
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: 1.4] [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
|
43
|
Akoumianakis I, Akawi N, Antoniades C. Exploring the Crosstalk between Adipose Tissue and the Cardiovascular System. Korean Circ J 2017; 47:670-685. [PMID: 28955384 PMCID: PMC5614942 DOI: 10.4070/kcj.2017.0041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 04/04/2017] [Indexed: 12/28/2022] Open
Abstract
Obesity is a clinical entity critically involved in the development and progression of cardiovascular disease (CVD), which is characterised by variable expansion of adipose tissue (AT) mass across the body as well as by phenotypic alterations in AT. AT is able to secrete a diverse spectrum of biologically active substances called adipocytokines, which reach the cardiovascular system via both endocrine and paracrine routes, potentially regulating a variety of physiological and pathophysiological responses in the vasculature and heart. Such responses include regulation of inflammation and oxidative stress as well as cell proliferation, migration and hypertrophy. Furthermore, clinical observations such as the “obesity paradox,” namely the fact that moderately obese patients with CVD have favourable clinical outcome, strongly indicate that the biological “quality” of AT may be far more crucial than its overall mass in the regulation of CVD pathogenesis. In this work, we describe the anatomical and biological diversity of AT in health and metabolic disease; we next explore its association with CVD and, importantly, novel evidence for its dynamic crosstalk with the cardiovascular system, which could regulate CVD pathogenesis.
Collapse
Affiliation(s)
- Ioannis Akoumianakis
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | - Nadia Akawi
- Division of Cardiovascular Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
44
|
Hartwig V, Guiducci L, Marinelli M, Pistoia L, Tegrimi TM, Iervasi G, Quinones-Galvan A, L'Abbate A. Multimodal Imaging for the Detection of Brown Adipose Tissue Activation in Women: A Pilot Study Using NIRS and Infrared Thermography. JOURNAL OF HEALTHCARE ENGINEERING 2017; 2017:5986452. [PMID: 29104746 PMCID: PMC5618764 DOI: 10.1155/2017/5986452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Revised: 06/26/2017] [Accepted: 08/10/2017] [Indexed: 01/06/2023]
Abstract
Purpose A clear link between obesity and brown adipose tissue (BAT) dysfunction has been recently demonstrated. The purpose of this pilot study is to determine if near-infrared spectroscopy (NIRS) 2D imaging together with infrared thermography (IRT) is capable of identifying thermal and vascular response in the supraclavicular (SCV) areas after the ingestion of an oral glucose load as a thermogenic stimulation. Method We studied two groups of women (obese versus lean) for discerning their different responses. NIRS and IRT images were acquired on the neck in the left SCV region during a 3 h oral glucose tolerance test (OGTT) and immediately after a cold stimulation. Results We detected a significant thermal response of BAT in SCV fossa in both groups. Both during OGTT and after cold stimulation, skin temperature was persistently higher in lean versus obese. This response was not coupled with changes in oxygen saturation of subcutaneous tissue in that area. Discussion and Conclusion The results show that NIRS/IRT may be a novel, noninvasive, radiation-free, easy to use, and low-cost method for monitoring, during the standard clinical practice, the diet and pharmacological intervention which aims to stimulate BAT as a potential therapeutic target against obesity and diabetes.
Collapse
Affiliation(s)
- Valentina Hartwig
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124 Pisa, Italy
| | - Letizia Guiducci
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124 Pisa, Italy
| | - Martina Marinelli
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124 Pisa, Italy
| | - Laura Pistoia
- Fondazione G. Monasterio, CNR-Regione Toscana, Via G. Moruzzi 1, 56124 Pisa, Italy
| | | | - Giorgio Iervasi
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124 Pisa, Italy
| | | | - Antonio L'Abbate
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124 Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant'Anna, Piazza Martiri della Libertà 33, 56127 Pisa, Italy
| |
Collapse
|
45
|
Palmer BF, Clegg DJ. Non-shivering thermogenesis as a mechanism to facilitate sustainable weight loss. Obes Rev 2017; 18:819-831. [PMID: 28547916 DOI: 10.1111/obr.12563] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 01/01/2023]
Abstract
Currently, there is a significant percentage of the population who are or will be classified as obese, necessitating novel strategies to facilitate sustainable weight loss. Reductions in basal metabolic rate occur in the face of weight loss and pose formidable barriers to individuals attempting to sustain meaningful weight reductions. Here, we discuss the mechanisms by which non-shivering thermogenesis may provide insight into metabolic pathways that can become druggable targets to facilitate sustainable weight loss. Specifically, we highlight the fact that non-shivering thermogenesis results in activation and expansion of brown and beige adipose tissues as well as activates pathways in skeletal muscle which increase metabolic flux and activity of muscle fibres through futile calcium cycling across the endoplasmic reticulum all facilitating an increase in metabolism. Finally, we highlight the fact there are sexual dimorphisms with respect to these metabolic processes in keeping with the National Institutes of Health mandate of treating sex as a biologic variable.
Collapse
Affiliation(s)
- B F Palmer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - D J Clegg
- Biomedical Research Department, Diabetes and Obesity Research Division, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
46
|
Bauters D, Cobbaut M, Geys L, Van Lint J, Hemmeryckx B, Lijnen HR. Loss of ADAMTS5 enhances brown adipose tissue mass and promotes browning of white adipose tissue via CREB signaling. Mol Metab 2017; 6:715-724. [PMID: 28702327 PMCID: PMC5485238 DOI: 10.1016/j.molmet.2017.05.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 12/24/2022] Open
Abstract
Objective A potential strategy to treat obesity – and the associated metabolic consequences – is to increase energy expenditure. This could be achieved by stimulating thermogenesis through activation of brown adipose tissue (BAT) and/or the induction of browning of white adipose tissue (WAT). Over the last years, it has become clear that several metalloproteinases play an important role in adipocyte biology. Here, we investigated the potential role of ADAMTS5. Methods Mice deficient in ADAMTS5 (Adamts5−/−) and wild-type (Adamts5+/+) littermates were kept on a standard of Western-type diet for 15 weeks. Energy expenditure and heat production was followed by indirect calorimetry. To activate thermogenesis, mice were treated with the β3-adrenergic receptor (β3-AR) agonist CL-316,243 or alternatively, exposed to cold for 2 weeks. Results Compared to Adamts5+/+ mice, Adamts5−/− mice have significantly more interscapular BAT and marked browning of their subcutaneous (SC) WAT. Thermogenic pathway analysis indicated, in the absence of ADAMTS5, enhanced β3-AR signaling via activation of the cAMP response element-binding protein (CREB). Additional β3-AR stimulation with CL-316,243 promoted browning of WAT in Adamts5+/+ mice but had no additive effect in Adamts5−/− mice. However, cold exposure induced more pronounced browning of WAT in Adamts5−/− mice. Conclusions These data indicate that ADAMTS5 plays a functional role in development of BAT and browning of WAT. Hence, selective targeting of ADAMTS5 could provide a novel therapeutic strategy for treatment/prevention of obesity and metabolic diseases. Mice deficient in ADAMTS5 have elevated interscapular brown adipose tissue mass. ADAMTS5 deficient mice show increased browning of their white adipose tissue. The thermogenic profile is enhanced via adrenergic signaling and CREB activation. ADAMTS5 seems an attractive therapeutic target for metabolic diseases.
Collapse
Key Words
- %ID/g, percentage injected dose per gram
- ADAMTS, A disintesgrin and metalloproteinase with a thrombospondin type-1 motif
- ADAMTS5
- AT, adipose tissue
- BAT, brown adipose tissue
- Beige
- Brown adipose tissue
- Browning
- CREB, cAMP responsive element-binding protein
- ECM, extracellular matrix
- GON, gonadal
- HFD, high-fat diet
- Obesity
- SC, subcutaneous
- SUV, standardized uptake value
- TLG, total lesion glycolysis
- Thermogenesis
- UCP1, uncoupling protein 1
- WAT, white adipose tissue
- β3-AR, beta-3 adrenergic receptor
Collapse
Affiliation(s)
- Dries Bauters
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, B-3000 Leuven, Belgium
| | - Mathias Cobbaut
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, B-3000 Leuven, Belgium
| | - Lotte Geys
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, B-3000 Leuven, Belgium
| | - Johan Van Lint
- Department of Cellular and Molecular Medicine, Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, B-3000 Leuven, Belgium
| | - Bianca Hemmeryckx
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, B-3000 Leuven, Belgium
| | - H Roger Lijnen
- Department of Cardiovascular Sciences, Center for Molecular and Vascular Biology, KU Leuven, B-3000 Leuven, Belgium
| |
Collapse
|
47
|
Singh R, Braga M, Reddy ST, Lee SJ, Parveen M, Grijalva V, Vergnes L, Pervin S. Follistatin Targets Distinct Pathways To Promote Brown Adipocyte Characteristics in Brown and White Adipose Tissues. Endocrinology 2017; 158:1217-1230. [PMID: 28324027 PMCID: PMC5460830 DOI: 10.1210/en.2016-1607] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022]
Abstract
We previously demonstrated that Fst expression is highest in brown adipose tissue (BAT) and skeletal muscle, but is also present at substantial levels in epididymal and subcutaneous white adipose tissues (WATs). Fst promotes mouse brown preadipocyte differentiation and promotes browning during differentiation of mouse embryonic fibroblasts. Fst-transgenic (Fst-Tg) mice show substantial increases in circulating Fst levels and increased brown adipose mass. BAT of Fst-Tg mice had increased expression of brown adipose-associated markers including uncoupling protein 1 (UCP1), PRDM16, PGC-1α, and Glut4. WATs from Fst-Tg mice show upregulation of brown/beige adipose markers and significantly increased levels of phosphorylated p38 MAPK/ERK1/2 proteins compared with the wild-type (WT) mice. Pharmacological inhibition of pp38 MAPK/pERK1/2 pathway of recombinant mouse Fst (rFst) treated differentiating 3T3-L1 cells led to significant blockade of Fst-induced UCP1 protein expression. On the other hand, BAT from Fst-Tg mice or differentiating mouse BAT cells treated with rFst show dramatic increase in Myf5 protein levels as well as upregulation of Zic1 and Lhx8 gene expression. Myf5 levels were significantly downregulated in Fst knock-out embryos and small inhibitory RNA-mediated inhibition of Myf5 led to significant inhibition of UCP1, Lhx8, and Zic1 gene expression and significant blockade of Fst-induced induction of UCP1 protein expression in mouse BAT cells. Both interscapular BAT and WAT tissues from Fst-Tg mice display enhanced response to CL316,243 treatment and decreased expression of pSmad3 compared with the WT mice. Therefore, our results indicate that Fst promotes brown adipocyte characteristics in both WAT and BAT depots in vivo through distinct mechanisms.
Collapse
MESH Headings
- 3T3-L1 Cells
- Adipocytes, Brown/physiology
- Adipocytes, White/physiology
- Adipose Tissue, Brown/anatomy & histology
- Adipose Tissue, Brown/physiology
- Adipose Tissue, White/anatomy & histology
- Adipose Tissue, White/physiology
- Animals
- Cell Differentiation/genetics
- Cell Transdifferentiation/genetics
- Cells, Cultured
- Embryo, Mammalian
- Female
- Follistatin/blood
- Follistatin/genetics
- Follistatin/physiology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Signal Transduction/genetics
- Thermogenesis/genetics
Collapse
Affiliation(s)
- Rajan Singh
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
- 2Obstetrics and Gynecology and
| | - Melissa Braga
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
| | - Srinivasa T. Reddy
- 2Obstetrics and Gynecology and
- Medicine, Molecular and Medical Pharmacology and
| | - Se-Jin Lee
- Johns Hopkins University School of Medicine, Department of Molecular Biology and Genetics, Baltimore, Maryland 21205
| | - Meher Parveen
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
| | | | - Laurent Vergnes
- Molecular Biology and Genetics, David Geffen School of Medicine at UCLA, Los Angeles, California 90095
| | - Shehla Pervin
- Division of Endocrinology and Metabolism, Charles R. Drew University of Medicine and Science, Los Angeles, California 90059; Departments of
- 2Obstetrics and Gynecology and
| |
Collapse
|
48
|
Pandit R, Beerens S, Adan RAH. Role of leptin in energy expenditure: the hypothalamic perspective. Am J Physiol Regul Integr Comp Physiol 2017; 312:R938-R947. [PMID: 28356295 DOI: 10.1152/ajpregu.00045.2016] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/27/2017] [Accepted: 03/28/2017] [Indexed: 01/08/2023]
Abstract
The adipocyte-derived hormone leptin is a peripheral signal that informs the brain about the metabolic status of an organism. Although traditionally viewed as an appetite-suppressing hormone, studies in the past decade have highlighted the role of leptin in energy expenditure. Leptin has been shown to increase energy expenditure in particular through its effects on the cardiovascular system and brown adipose tissue (BAT) thermogenesis via the hypothalamus. The current review summarizes the role of leptin signaling in various hypothalamic nuclei and its effects on the sympathetic nervous system to influence blood pressure, heart rate, and BAT thermogenesis. Specifically, the role of leptin signaling on three different hypothalamic nuclei, the dorsomedial hypothalamus, the ventromedial hypothalamus, and the arcuate nucleus, is reviewed. It is known that all of these brain regions influence the sympathetic nervous system activity and thereby regulate BAT thermogenesis and the cardiovascular system. Thus the current work focuses on how leptin signaling in specific neuronal populations within these hypothalamic nuclei influences certain aspects of energy expenditure.
Collapse
Affiliation(s)
- R Pandit
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - S Beerens
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - R A H Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
49
|
Darcy J, McFadden S, Fang Y, Huber JA, Zhang C, Sun LY, Bartke A. Brown Adipose Tissue Function Is Enhanced in Long-Lived, Male Ames Dwarf Mice. Endocrinology 2016; 157:4744-4753. [PMID: 27740871 PMCID: PMC5133358 DOI: 10.1210/en.2016-1593] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ames dwarf mice (Prop1df/df) are long-lived due to a loss of function mutation, resulting in deficiency of GH, TSH, and prolactin. Along with a marked extension of longevity, Ames dwarf mice have improved energy metabolism as measured by an increase in their oxygen consumption and heat production, as well as a decrease in their respiratory quotient. Along with alterations in energy metabolism, Ames dwarf mice have a lower core body temperature. Moreover, Ames dwarf mice have functionally altered epididymal white adipose tissue (WAT) that improves, rather than impairs, their insulin sensitivity due to a shift from pro- to anti-inflammatory cytokine secretion. Given the unique phenotype of Ames dwarf epididymal WAT, their improved energy metabolism, and lower core body temperature, we hypothesized that Ames dwarf brown adipose tissue (BAT) may function differently from that of their normal littermates. Here we use histology and RT-PCR to demonstrate that Ames dwarf mice have enhanced BAT function. We also use interscapular BAT removal to demonstrate that BAT is necessary for Ames dwarf energy metabolism and thermogenesis, whereas it is less important for their normal littermates. Furthermore, we show that Ames dwarf mice are able to compensate for loss of interscapular BAT by using their WAT depots as an energy source. These findings demonstrate enhanced BAT function in animals with GH and thyroid hormone deficiencies, chronic reduction of body temperature, and remarkably extended longevity.
Collapse
Affiliation(s)
- Justin Darcy
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Samuel McFadden
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Yimin Fang
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Joshua A Huber
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Chi Zhang
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Liou Y Sun
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Andrzej Bartke
- Department of Internal Medicine (J.D., S.M., Y.F., J.A.H., C.Z., A.B.), Geriatric Research, and Department of Medical Microbiology, Immunology, and Cell Biology (J.D., A.B.), Southern Illinois University School of Medicine, Springfield, Illinois 62702; Institute of Cardiovascular Disease (C.Z.), Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang 421001, People's Republic of China; and Department of Biology (L.Y.S.), University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
50
|
Caron A, Richard D. Neuronal systems and circuits involved in the control of food intake and adaptive thermogenesis. Ann N Y Acad Sci 2016; 1391:35-53. [PMID: 27768821 DOI: 10.1111/nyas.13263] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 08/18/2016] [Accepted: 08/24/2016] [Indexed: 12/27/2022]
Abstract
With the still-growing prevalence of obesity worldwide, major efforts are made to understand the various behavioral, environmental, and genetic factors that promote excess fat gain. Obesity results from an imbalance between energy intake and energy expenditure, which emphasizes the importance of deciphering the mechanisms behind energy balance regulation to understand its physiopathology. The control of energy balance is assured by brain systems/circuits capable of generating adequate ingestive and thermogenic responses to maintain the stability of energy reserves, which implies a proper integration of the homeostatic signals that inform about the status of the energy stores. In this article, we overview the organization and functionality of key neuronal circuits or pathways involved in the control of food intake and energy expenditure. We review the role of the corticolimbic (executive and reward) and autonomic systems that integrate their activities to regulate energy balance. We also describe the mechanisms and pathways whereby homeostatic sensing is achieved in response to variations of homeostatic hormones, such as leptin, insulin, and ghrelin, while putting some emphasis on the prominent importance of the mechanistic target of the rapamycin signaling pathway in coordinating the homeostatic sensing process.
Collapse
Affiliation(s)
- Alexandre Caron
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec and Faculty of Medicine, Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| | - Denis Richard
- Institut Universitaire de Cardiologie et de Pneumologie de Quebec and Faculty of Medicine, Department of Medicine, Université Laval, Quebec City, Quebec, Canada
| |
Collapse
|