1
|
Lee AH, Orliaguet L, Youm YH, Maeda R, Dlugos T, Lei Y, Coman D, Shchukina I, Andhey PS, Smith SR, Ravussin E, Stadler K, Chen B, Artyomov MN, Hyder F, Horvath TL, Schneeberger M, Sugiura Y, Dixit VD. Cysteine depletion triggers adipose tissue thermogenesis and weight loss. Nat Metab 2025:10.1038/s42255-025-01297-8. [PMID: 40461845 DOI: 10.1038/s42255-025-01297-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/09/2025] [Indexed: 06/11/2025]
Abstract
Caloric restriction and methionine restriction-driven enhanced lifespan and healthspan induces 'browning' of white adipose tissue, a metabolic response that increases heat production to defend core body temperature. However, how specific dietary amino acids control adipose thermogenesis is unknown. Here, we identified that weight loss induced by caloric restriction in humans reduces thiol-containing sulfur amino acid cysteine in white adipose tissue. Systemic cysteine depletion in mice causes lethal weight loss with increased fat utilization and browning of adipocytes that is rescued upon restoration of cysteine in diet. Mechanistically, cysteine-restriction-induced adipose browning and weight loss requires sympathetic nervous system-derived noradrenaline signalling via β3-adrenergic-receptors that is independent of FGF21 and UCP1. In obese mice, cysteine deprivation induced rapid adipose browning, increased energy expenditure leading to 30% weight loss and reversed metabolic inflammation. These findings establish that cysteine is essential for organismal metabolism as removal of cysteine in the host triggers adipose browning and rapid weight loss.
Collapse
Affiliation(s)
- Aileen H Lee
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Lucie Orliaguet
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Yun-Hee Youm
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Rae Maeda
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tamara Dlugos
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Yuanjiu Lei
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, USA
| | - Irina Shchukina
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Prabhakar Sairam Andhey
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Bandy Chen
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University, New Haven, CT, USA
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA
- Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - Marc Schneeberger
- Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT, USA
| | - Yuki Sugiura
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Vishwa Deep Dixit
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
2
|
Gai Y, Ma G, Yang S, Hu Z, Ma Y, He R, Zhang Y, Huang S, Azzaz HH, Gu Z, Mao S, Ghaffari MH, Chen Y. Effects of maternal blood beta-hydroxybutyrate on brown adipose tissue functions and thermogenic and metabolic health in neonatal calves. J Dairy Sci 2025; 108:6439-6454. [PMID: 40222674 DOI: 10.3168/jds.2024-26123] [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: 12/07/2024] [Accepted: 03/12/2025] [Indexed: 04/15/2025]
Abstract
Maternal metabolic health, particularly during late pregnancy, plays a crucial role in fetal development and postnatal metabolic function. Elevated levels of BHB in dry cows, commonly observed in late gestation, may affect offspring development, but the effects on brown adipose tissue (BAT) and metabolic health remain unclear. In this study, 60 pregnant Holstein dairy cows were categorized into 2 groups based on serum BHB concentrations measured at 1, 3, 5, and 7 wk after dry-off: maternal low BHB (n = 30; mean ± SEM, 0.21 ± 0.005 mM) and maternal high BHB (n = 30; mean ± SEM, 0.64 ± 0.02 mM). Blood metabolites, including BHB, nonesterified fatty acids, and glucose, were monitored throughout the dry period. Calves born from these cows were evaluated for body growth, body temperature, glucose sensitivity, fecal, and cough score during the first month of life, with perirenal BAT and skin samples collected for analysis of thermogenic gene expression. Expression of stress genes, including CIRBP, HSP70, and HSBP1, was analyzed in skin tissue. Expression of thermogenic genes, including UCP-1, CREBP4, and CPT1B, and protein contents of UCP-1, PGC-1a were analyzed in BAT. In vitro, stromal vascular fractions were also isolated in calf's BAT, and further induced for brown adipocyte formation with dosed BHB supplementation. Results showed no differences in birth weight, body size, and body temperatures of calves born to maternal high BHB cows compared with calves born to maternal low BHB cows. However, the calves from the maternal high BHB group had higher expressions of stress genes in the skin, and decreased BAT mass and expression of thermogenic genes. Compared with the maternal low BHB group, 1-mo-old calves in the maternal high BHB group also showed significantly lower BAT mass, decreased expression of thermogenic genes, such as UCP-1, CREBP4, and CPT1B, and decreased mitochondrial density, indicating impaired BAT development. In addition, the calves from the maternal high BHB group showed reduced glucose sensitivity, as evidenced by their inability to maintain stable blood glucose levels during a glucose tolerance test. Protein concentrations of UCP-1 and PGC-1a were significantly lower in the BAT of calves born to maternal high BHB cows. In vitro, BHB supplementation inhibited brown adipocyte differentiation and thermogenesis, supporting the elevated maternal BHB impairs brown adipogenesis and mitochondrial biogenesis. Overall, this study demonstrates that calves born from elevated maternal BHB levels (∼0.64 mM) within the normal physiological range in the dry period significantly had impaired perinatal BAT development, thermogenesis, and glucose metabolism, highlighting the roles of maternal metabolic health in programming metabolic and thermoregulatory capacity in offspring.
Collapse
Affiliation(s)
- Yang Gai
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Guiling Ma
- College of Agro-Grassland Science, Nanjing Agricultural University, Nanjing 210095, China
| | - Shuyan Yang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Zhiyong Hu
- College of Animal Science, Shandong Agricultural University, Taian 21018, China
| | - Yulin Ma
- College of Agriculture, Ningxia University, Yinchuan 750021, China
| | - Rui He
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shilong Huang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Hossam H Azzaz
- Dairy Department National Research Center, Giza, Cairo 12622, Egypt
| | - Zhaobing Gu
- Faculty of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Shengyong Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Morteza H Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, 53115 Bonn, Germany
| | - Yanting Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; National Center for International Research on Animal Gut Nutrition, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, Nanjing Agricultural University, Nanjing 210095 China.
| |
Collapse
|
3
|
Sun Q, Cui X, Yin D, Li J, Li J, Du L. Molecular mechanisms of UCP1-independent thermogenesis: the role of futile cycles in energy dissipation. J Physiol Biochem 2025:10.1007/s13105-025-01090-x. [PMID: 40380026 DOI: 10.1007/s13105-025-01090-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 05/01/2025] [Indexed: 05/19/2025]
Abstract
Adipose tissue thermogenesis has emerged as a prominent research focus for the treatment of metabolic diseases, particularly through mitochondrial uncoupling, which oxidizes nutrients to produce heat rather than synthesizing ATP. Uncoupling protein 1 (UCP1) has garnered significant attention as a core protein mediating non-shivering thermogenesis(NST). However, recent studies indicate that energy dissipation can also occur via UCP1-independent thermogenesis, partially driven by futile metabolic cycles. These cycles involve ATP depletion coupled with reversible energy reactions, resulting in futile energy expenditure. Unlike classical UCP1-mediated thermogenesis, futile cycling is not confined to brown and beige adipose tissue, suggesting a broader range of therapeutic targets. These findings open new avenues for targeting these pathways to enhance metabolic health. This review explores the characteristics and distinctions of the primary metabolic organs (adipose tissue, liver, and skeletal muscle) involved in the futile cycles of thermogenesis. It further elaborates on the cellular and molecular mechanisms underlying calcium, creatine, and lipid cycling, emphasizing their strengths, limitations, and roles beyond thermogenesis.
Collapse
Affiliation(s)
- Quanhao Sun
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Xinyue Cui
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Dong Yin
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Juan Li
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Jiarui Li
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Likun Du
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
4
|
Sleiman L, Dinescu S. Role of Non-Coding RNAs in White and Brown Adipose Tissue Differentiation and Development. Noncoding RNA 2025; 11:30. [PMID: 40407588 PMCID: PMC12101253 DOI: 10.3390/ncrna11030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 04/18/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Abstract
Adipocyte differentiation is a complex process in which pluripotent mesenchymal stem cells (MSCs) differentiate and develop into mature fat cells, also known as adipocytes. This process is controlled by various transcription factors, hormones, and signaling molecules that regulate the development of these cells. Recently, an increasing number of non-coding RNAs (ncRNAs), especially microRNAs (miRNAs), have been established to be involved in the regulation of many biological processes, including adipocyte differentiation, development, metabolism, and energy homeostasis of white and brown adipose tissue. Several in vitro and in vivo studies reported the significant role of ncRNAs in either promoting or inhibiting adipocyte differentiation into white or brown fat cells by targeting specific transcription factors and regulating the expression of key adipogenic genes. Identifying the function of ncRNAs and their subsequent targets contributes to our understanding of how these molecules can be used as potential biomarkers and tools for therapies against obesity, diabetes, and other diseases related to obesity. This could also contribute to advancements in tissue-engineering based treatments. In this review, we intended to present an up-to-date comprehensive literature overview of the role of ncRNAs, including miRNAs, long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), focusing particularly on miRNAs, in regulating the differentiation and development of cells into white and brown adipose tissue. In addition, we further discuss the potential use of these molecules as biomarkers for the development of novel therapeutic strategies for future personalized treatment options for patients.
Collapse
Affiliation(s)
- Lea Sleiman
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania;
- Research Institute of the University of Bucharest (ICUB), 050663 Bucharest, Romania
| |
Collapse
|
5
|
Bunk J, Hussain MF, Delgado-Martin M, Samborska B, Ersin M, Shaw A, Rahbani JF, Kazak L. The Futile Creatine Cycle powers UCP1-independent thermogenesis in classical BAT. Nat Commun 2025; 16:3221. [PMID: 40185737 PMCID: PMC11971250 DOI: 10.1038/s41467-025-58294-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 03/18/2025] [Indexed: 04/07/2025] Open
Abstract
Classical brown adipose tissue (BAT) is traditionally viewed as relying exclusively on uncoupling protein 1 (UCP1) for thermogenesis via inducible proton leak. However, the physiological significance of UCP1-independent mechanisms linking substrate oxidation to ATP turnover in classical BAT has remained unclear. Here, we identify the Futile Creatine Cycle (FCC), a mitochondrial-localized energy-wasting pathway involving creatine phosphorylation by creatine kinase b (CKB) and phosphocreatine hydrolysis by tissue-nonspecific alkaline phosphatase (TNAP), as a key UCP1-independent thermogenic mechanism in classical BAT. Reintroducing mitochondrial-targeted CKB exclusively into interscapular brown adipocytes in vivo restores thermogenesis and cold tolerance in mice lacking native UCP1 and CKB, in a TNAP-dependent manner. Furthermore, mice with inducible adipocyte-specific co-deletion of TNAP and UCP1 exhibit severe cold-intolerance. These findings challenge the view that BAT thermogenesis depends solely on UCP1 because of insufficient ATP synthase activity and establishes the FCC as a physiologically relevant thermogenic pathway in classical BAT.
Collapse
Affiliation(s)
- Jakub Bunk
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Mohammed F Hussain
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Maria Delgado-Martin
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Bozena Samborska
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Mina Ersin
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Abhirup Shaw
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Janane F Rahbani
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada.
- Department of Biochemistry, McGill University, Montreal, QC, Canada.
| |
Collapse
|
6
|
Auger C, Li M, Fujimoto M, Ikeda K, Yook JS, O'Leary TR, Caycedo MPH, Xiaohan C, Oikawa S, Verkerke ARP, Shinoda K, Griffin PR, Inaba K, Stimson RH, Kajimura S. Identification of a molecular resistor that controls UCP1-independent Ca 2+ cycling thermogenesis in adipose tissue. Cell Metab 2025:S1550-4131(25)00112-3. [PMID: 40199326 DOI: 10.1016/j.cmet.2025.03.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 01/07/2025] [Accepted: 03/11/2025] [Indexed: 04/10/2025]
Abstract
Adipose tissue thermogenesis contributes to energy balance via mitochondrial uncoupling protein 1 (UCP1) and UCP1-independent pathways. Among UCP1-independent thermogenic mechanisms, one involves Ca2+ cycling via SERCA2b in adipose tissue; however, the underlying molecular basis remains elusive. Here, we report that an endoplasmic reticulum (ER) membrane-anchored peptide, C4orf3 (also known as another regulin [ALN]), uncouples SERCA2b Ca2+ transport from its ATP hydrolysis, rendering the SERCA2b-C4orf3 complex exothermic. Loss of C4orf3/ALN improved the energetic efficiency of SERCA2b-dependent Ca2+ transport without affecting SERCA2 expression, thereby reducing adipose tissue thermogenesis and increasing the adiposity of mice. Notably, genetic depletion of C4orf3 resulted in compensatory activation of UCP1-dependent thermogenesis following cold challenge. We demonstrated that genetic loss of both C4orf3 and Ucp1 additively impaired cold tolerance in vivo. Together, this study identifies C4orf3 as the molecular resistor to SERCA2b-mediated Ca2+ import that plays a key role in UCP1-independent thermogenesis and energy balance.
Collapse
Affiliation(s)
- Christopher Auger
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Mark Li
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Masanori Fujimoto
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Kenji Ikeda
- Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University, Tokyo, Japan
| | - Jin-Seon Yook
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Timothy R O'Leary
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - María Paula Huertas Caycedo
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Cai Xiaohan
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Satoshi Oikawa
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Anthony R P Verkerke
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA
| | - Kosaku Shinoda
- Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, NY, USA
| | - Patrick R Griffin
- Department of Molecular Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Kenji Inaba
- Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan
| | - Roland H Stimson
- University/BHF Centre for Cardiovascular Science, University of Edinburgh, Queen's Medical Research Institute, Edinburgh, UK
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, and Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
7
|
Yuan B, Doxsey W, Tok Ö, Kwon YY, Liang Y, Inouye KE, Hotamışlıgil GS, Hui S. An organism-level quantitative flux model of energy metabolism in mice. Cell Metab 2025; 37:1012-1023.e6. [PMID: 39983714 PMCID: PMC11964847 DOI: 10.1016/j.cmet.2025.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 11/21/2024] [Accepted: 01/09/2025] [Indexed: 02/23/2025]
Abstract
Mammalian tissues feed on nutrients in the blood circulation. At the organism level, mammalian energy metabolism is comprised of the oxidation, storage, interconversion, and release of circulating nutrients. Here, by integrating isotope tracer infusion, mass spectrometry, and isotope gas analyzer measurement, we developed a framework to systematically quantify fluxes through these metabolic processes for 10 major circulating energy nutrients in mice, resulting in an organism-level quantitative flux model of energy metabolism. This model revealed in wild-type mice that circulating nutrients have metabolic cycling fluxes dominant to their oxidation fluxes, with distinct partitions between cycling and oxidation for individual circulating nutrients. Applications of this framework in obese mouse models showed extensive elevation of metabolic cycling fluxes in ob/ob mice but not in diet-induced obese mice on a per-animal or per-lean mass basis. Our framework is a valuable tool to reveal new features of energy metabolism in physiological and disease conditions.
Collapse
Affiliation(s)
- Bo Yuan
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Will Doxsey
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Özlem Tok
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Young-Yon Kwon
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Yanshan Liang
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Karen E Inouye
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Gökhan S Hotamışlıgil
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Sheng Hui
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
8
|
Ranea-Robles P, Lund C, Svendsen C, Gil C, Lund J, Kleinert M, Clemmensen C. Time-Resolved Effects of Short-term Overfeeding on Energy Balance in Mice. Diabetes 2025; 74:502-513. [PMID: 39787442 PMCID: PMC11926271 DOI: 10.2337/db24-0289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
ARTICLE HIGHLIGHTS Intragastric overfeeding reveals insights into the homeostatic recovery from experimental weight gain. Protection against short-term, overfeeding-induced weight gain primarily involves a profound reduction in food intake and possibly an adaptive increase in energy expenditure. UCP1-mediated thermogenesis is not essential for homeostatic protection against short-term, overfeeding-induced weight gain.
Collapse
Affiliation(s)
- Pablo Ranea-Robles
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
- Departamento de Fisiología, Facultad de Medicina, Universidad de Granada, Granada, Spain
- Centro de Investigación Biomédica, Universidad de Granada, Granada, Spain
- Instituto de Investigación Biosanitaria, Granada, Spain
| | - Camilla Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Charlotte Svendsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Cláudia Gil
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Maximilian Kleinert
- German Center for Diabetes Research (DZD), Munich-Neuherberg, Germany
- Department of Molecular Physiology of Exercise and Nutrition, German Institute of Human Nutrition, Potsdam-Rehbruecke, Nuthetal, Germany
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
9
|
Yi T, Wu S, Yang Y, Li X, Yang S, Zhang Y, Zhang L, Hu Y, Zhang G, Li J, Yang D. Single-nucleus RNA sequencing reveals dynamic changes in the microenvironment of visceral adipose tissue and metabolic characteristics after cold exposure. Front Endocrinol (Lausanne) 2025; 16:1562431. [PMID: 40196457 PMCID: PMC11973077 DOI: 10.3389/fendo.2025.1562431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Visceral adipose tissue (VAT) plays a crucial role in regulating systemic metabolic balance. Excess accumulation of VAT is closely associated with various metabolic disorders, a process that involves the coordinated actions of multiple cell types within the tissue. Cold exposure, as a potential intervention, has been proposed to improve metabolic dysfunction. However, the heterogeneity of VAT and its comprehensive metabolic characteristics under cold exposure remain unclear. Methods We collected epididymal white adipose tissue (eWAT) of C57BL/6J mice after cold exposure at three different time points for single-nucleus RNA sequencing (snRNA-seq) analysis. Results We successfully identified ten major cell types in eWAT, enabling understanding of the dynamic changes in the eWAT microenvironment and its metabolic features during cold exposure. This study revealed that cold exposure for 1 day reduced cellular metabolic activity and intercellular communication in eWAT including receptor-ligand-based cell communication and metabolite-mediated interactions. However, after 14 days of cold acclimation, the metabolic activity of adipocytes was significantly enhanced, and intercellular metabolic communication was restored. Additionally, prolonged cold exposure promoted the formation of a distinct adipocyte subpopulation that may be associated with UCP1-independent thermogenesis. These changes may be a new homeostatic state established by VAT to adapt to the cold environment. The study also identified the importance of adipocytes, adipose stem and progenitor cells, myeloid cells, and endothelial cells in the process of cold adaptation. Discussion This research provides valuable insights into the cellular heterogeneity, adipocyte remodeling, and metabolic reprogramming in eWAT after cold exposure. It highlights the critical role of transcriptional dynamics in eWAT during cold exposure and provides new perspectives on the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Ting Yi
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuai Wu
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yusha Yang
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Xi Li
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Shuran Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yongqiang Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Li Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Yuyu Hu
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Guanyu Zhang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| | - Jun Li
- School of Public Health, The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, China
| | - Danfeng Yang
- Academy of Military Medical Sciences, Academy of Military Sciences, Tianjin, China
| |
Collapse
|
10
|
Rodríguez-Díaz A, Diéguez C, López M, Freire-Agulleiro Ó. FAcTs on fire: Exploring thermogenesis. ADVANCES IN GENETICS 2025; 113:172-198. [PMID: 40409797 DOI: 10.1016/bs.adgen.2025.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2025]
Abstract
Thermoregulation is a fundamental biological process that allows birds and mammals to maintain a stable internal temperature despite environmental fluctuations, a mechanism shaped by millions of years of evolution. Non-shivering thermogenesis (NST), primarily driven by brown adipose tissue (BAT), plays a central role in thermoregulation by not only helping maintain energy homeostasis but also influencing broader metabolic and physiological processes. Recent research has revealed that BAT thermogenesis is regulated by peripheral hormones and at a central level, with key hypothalamic energy-sensing pathways-such as AMP-activated protein kinase (AMPK) and endoplasmic reticulum (ER) stress-playing critical roles. Beyond its metabolic functions, BAT and NST have emerged as important contributors to tumor biology, offering novel therapeutic strategies for metabolic and oncological diseases. This review explores the intricate mechanisms underpinning NST, including UCP1-dependent thermogenesis and alternative pathways such as creatine cycling, calcium-dependent thermogenesis, and lipid cycling. Emerging evidence further highlights BAT's potential in to modulate tumor metabolism, with pharmacological and genetic approaches showing promise in reshaping the tumor microenvironment. This growing body of knowledge offers exciting prospects for targeting BAT thermogenesis in treating obesity and other metabolic diseases.
Collapse
Affiliation(s)
- Amanda Rodríguez-Díaz
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain
| | - Óscar Freire-Agulleiro
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Madrid, Spain.
| |
Collapse
|
11
|
Festuccia WT. mTORC1 and 2 Adrenergic Regulation and Function in Brown Adipose Tissue. Physiology (Bethesda) 2025; 40:0. [PMID: 39470603 DOI: 10.1152/physiol.00023.2024] [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: 05/07/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 10/30/2024] Open
Abstract
Brown adipose tissue (BAT) thermogenesis results from the uncoupling of mitochondrial inner membrane proton gradient mediated by uncoupling protein 1 (UCP-1), which is activated by lipolysis-derived fatty acids. Norepinephrine (NE) secreted by sympathetic innervation not only activates BAT lipolysis and UCP-1 but, uniquely in brown adipocytes, promotes "futile" metabolic cycles and enhances BAT thermogenic capacity by increasing UCP-1 content, mitochondrial biogenesis, and brown adipocyte hyperplasia. NE exerts these actions by triggering signaling in the canonical G protein-coupled β-adrenergic receptors, cAMP, and protein kinase A (PKA) pathway, which in brown adipocytes is under a complex and intricate cross talk with important growth-promoting signaling pathways such as those of mechanistic target of rapamycin (mTOR) complexes 1 (mTORC1) and 2 (mTORC2). This article reviews evidence suggesting that mTOR complexes are modulated by and participate in the thermogenic, metabolic, and growth-promoting effects elicited by NE in BAT and discusses current gaps and future directions in this field of research.
Collapse
|
12
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|
13
|
Shabalina IG, Jiménez B, Sousa-Filho CPB, Cannon B, Nedergaard J. In isolated brown adipose tissue mitochondria, UCP1 is not essential for - nor involved in - the uncoupling effects of the classical uncouplers FCCP and DNP. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2025; 1866:149516. [PMID: 39357779 DOI: 10.1016/j.bbabio.2024.149516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/31/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Recent patch-clamp studies of mitoplasts have challenged the traditional view that classical chemical uncoupling (by e.g. FCCP or DNP) is due to the protonophoric property of these substances themselves. These studies instead suggest that in brown-fat mitochondria, FCCP- and DNP-induced uncoupling is mediated through activation of UCP1 (and in other tissues by activation of the adenine nucleotide transporter). These studies thus advocate an entirely new paradigm for the interpretation of standard bioenergetic experiments. To examine whether these patch-clamp results obtained in brown-fat mitoplasts are directly transferable to classical isolated brown-fat mitochondria studies, we investigated the effects of FCCP and DNP in brown-fat mitochondria from wildtype and UCP1 KO mice, comparing the FCCP and DNP effects with those of a fatty acid (oleate), a bona fide activator of UCP1. Whereas the sensitivity of brown-fat mitochondria to oleate was much higher in UCP1-containing than in UCP1 KO mitochondria, there was no difference in sensitivity to FCCP and DNP between these mitochondria, neither in oxygen consumption rate nor in membrane potential studies. Correspondingly, the UCP1-dependent ability of GDP to competitively inhibit activation by oleate was not seen with FCCP and DNP. It would thus be premature to abandon the established bioenergetic interpretation of chemical uncoupler effects in classical isolated brown-fat mitochondria-and probably also generally in this type of mitochondrial study. Understanding the molecular and structural reasons for the different outcomes of mitoplast and mitochondrial studies is a challenging task.
Collapse
Affiliation(s)
- Irina G Shabalina
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Sweden
| | - Beatriz Jiménez
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Sweden
| | | | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Sweden.
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91, Sweden
| |
Collapse
|
14
|
Chang L, Meng F, Jiao B, Zhou T, Su R, Zhu C, Wu Y, Ling Y, Wang S, Wu K, Zhang D, Cao J. Integrated analysis of omics reveals the role of scapular fat in thermogenesis adaptation in sunite sheep. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY. PART D, GENOMICS & PROTEOMICS 2024; 52:101292. [PMID: 39018792 DOI: 10.1016/j.cbd.2024.101292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/19/2024]
Abstract
Inhabiting some of the world's most inhospitable climatic regions, the Sunite Mongolian sheep generates average temperatures as low as 4.3 °C and a minimum temperature of -38.8 °C; in these environments, they make essential cold adaptations. In this regard, scapular fat tissues from Mongolian sheep were collected both in winter and summer for transcriptomic and proteomic analyses to identify genes related to adaptive thermogenesis. In the transcriptome analysis, 588 differentially expressed genes were identified to participate in smooth muscle activity and fat metabolism, as well as in nutrient regulation. There were 343 upregulated and 245 downregulated genes. GO and KEGG pathway analyses on these genes revealed their participation in regulating smooth muscle activity, metabolism of fats, and nutrients. Proteomic analysis showed the differential expression of 925 proteins: among them, there are 432 up- and 493 down-expressed proteins. These proteins are mainly involved in oxidative phosphorylation, respiratory chain complex assembly, and ATP production by electron transport. Furthermore, using both sets at a more detailed level of analysis revealed over-representation in gene ontology categories related to hormone signaling, metabolism of lipids, the pentose phosphate pathway, the TCA cycle, and especially the process of oxidative phosphorylation. The identified essential genes and proteins were further validated by quantitative real-time polymerase chain reaction and Western blotting, respectively; key metabolic network constriction was constructed. The present study emphasized the critical role of lipid turnover in scapular fat for thermogenic adaptation in Sunite sheep.
Collapse
Affiliation(s)
- Longwei Chang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Fanhua Meng
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| | - Boran Jiao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Tong Zhou
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Rina Su
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Chunxiao Zhu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Yi Wu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Yu Ling
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Shenyuan Wang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Kaifeng Wu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China
| | - Dong Zhang
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| | - Junwei Cao
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China; Inner Mongolia Autonomous Region Key Laboratory of Biomanufacturing, Hohhot 010018, China.
| |
Collapse
|
15
|
Lai TH, Hwang JS, Ngo QN, Lee DK, Kim HJ, Kim DR. A comparative assessment of reference genes in mouse brown adipocyte differentiation and thermogenesis in vitro. Adipocyte 2024; 13:2330355. [PMID: 38527945 PMCID: PMC10965104 DOI: 10.1080/21623945.2024.2330355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 03/06/2024] [Indexed: 03/27/2024] Open
Abstract
Adipogenic differentiation and thermogenesis in brown adipose tissue (BAT) undergo dynamic processes, altering phenotypes and gene expressions. Proper reference genes in gene expression analysis are crucial to mitigate experimental variances and ensure PCR efficacy. Unreliable reference genes can lead to erroneous gene expression quantification, resulting in data misinterpretation. This study focused on identifying suitable reference genes for mouse brown adipocyte research, utilizing brown adipocytes from the Ucp1-luciferase ThermoMouse model. Comparative analysis of gene expression data under adipogenesis and thermogenesis conditions was conducted, validating 13 housekeeping genes through various algorithms, including DeltaCq, BestKeeper, geNorm, Normfinder, and RefFinder. Tbp and Rer1 emerged as optimal references for Ucp1 and Pparg expression in brown adipogenesis, while Tbp and Ubc were ideal for the expression analysis of these target genes in thermogenesis. Conversely, certain conventional references, including Actb, Tubb5, and Gapdh, proved unstable as reference genes under both conditions. These findings stress the critical consideration of reference gene selection in gene expression analysis within specific biological systems to ensure accurate conclusions.
Collapse
Affiliation(s)
- Trang Huyen Lai
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Jin Seok Hwang
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Quang Nhat Ngo
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Dong-Kun Lee
- Department of Physiology and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Hyun Joon Kim
- Department of Anatomy and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| | - Deok Ryong Kim
- Department of Biochemistry and Convergence Medical Sciences and Institute of Medical Science, Gyeongsang National University, College of Medicine, Jinju, South Korea
| |
Collapse
|
16
|
Natarajan D, Plakkot B, Tiwari K, Ekambaram S, Wang W, Rudolph M, Mohammad MA, Chacko SK, Subramanian M, Tarantini S, Yabluchanskiy A, Ungvari Z, Csiszar A, Balasubramanian P. Chronic β3-AR stimulation activates distinct thermogenic mechanisms in brown and white adipose tissue and improves systemic metabolism in aged mice. Aging Cell 2024; 23:e14321. [PMID: 39177077 PMCID: PMC11634714 DOI: 10.1111/acel.14321] [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: 05/28/2024] [Revised: 07/30/2024] [Accepted: 08/03/2024] [Indexed: 08/24/2024] Open
Abstract
Adipose thermogenesis has been actively investigated as a therapeutic target for improving metabolic dysfunction in obesity. However, its applicability to middle-aged and older populations, which bear the highest obesity prevalence in the United States (approximately 40%), remains uncertain due to age-related decline in thermogenic responses. In this study, we investigated the effects of chronic thermogenic stimulation using the β3-adrenergic (AR) agonist CL316,243 (CL) on systemic metabolism and adipose function in aged (18-month-old) C57BL/6JN mice. Sustained β3-AR treatment resulted in reduced fat mass, increased energy expenditure, increased fatty acid oxidation and mitochondrial activity in adipose depots, improved glucose homeostasis, and a favorable adipokine profile. At the cellular level, CL treatment increased uncoupling protein 1 (UCP1)-dependent thermogenesis in brown adipose tissue (BAT). However, in white adipose tissue (WAT) depots, CL treatment increased glycerol and lipid de novo lipogenesis (DNL) and turnover suggesting the activation of the futile substrate cycle of lipolysis and reesterification in a UCP1-independent manner. Increased lipid turnover was also associated with the simultaneous upregulation of proteins involved in glycerol metabolism, fatty acid oxidation, and reesterification in WAT. Further, a dose-dependent impact of CL treatment on inflammation was observed, particularly in subcutaneous WAT, suggesting a potential mismatch between fatty acid supply and oxidation. These findings indicate that chronic β3-AR stimulation activates distinct cellular mechanisms that increase energy expenditure in BAT and WAT to improve systemic metabolism in aged mice. Considering that people lose BAT with aging, activation of futile lipid cycling in WAT presents a novel strategy for improving age-related metabolic dysfunction.
Collapse
Affiliation(s)
- Duraipandy Natarajan
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Bhuvana Plakkot
- Department of Physiological Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Kritika Tiwari
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Shoba Ekambaram
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Weidong Wang
- Department of MedicineUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Michael Rudolph
- Department of Biochemistry and Physiology and Harold Hamm Diabetes CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Mahmoud A. Mohammad
- USDA/ARS Children's Nutrition Research CenterBaylor College of MedicineHoustonTexasUSA
| | - Shaji K. Chacko
- USDA/ARS Children's Nutrition Research CenterBaylor College of MedicineHoustonTexasUSA
| | - Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary MedicineOklahoma State UniversityStillwaterOklahomaUSA
| | - Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public HealthSemmelweis UniversityBudapestHungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Department of Health Promotion Sciences, College of Public HealthUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- USDA/ARS Children's Nutrition Research CenterBaylor College of MedicineHoustonTexasUSA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public HealthSemmelweis UniversityBudapestHungary
| | - Anna Csiszar
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public HealthSemmelweis UniversityBudapestHungary
| | - Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration, and Healthy Brain Aging Program, Department of NeurosurgeryUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Oklahoma Center for Geroscience and Healthy Brain AgingUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- The Peggy and Charles Stephenson Cancer CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
- Department of Biochemistry and Physiology and Harold Hamm Diabetes CenterUniversity of Oklahoma Health Sciences CenterOklahoma CityOklahomaUSA
| |
Collapse
|
17
|
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
|
18
|
Zhai X, Dang L, Wang S, Sun C. The SIRT5-Mediated Upregulation of C/EBPβ Promotes White Adipose Tissue Browning by Enhancing UCP1 Signaling. Int J Mol Sci 2024; 25:10514. [PMID: 39408844 PMCID: PMC11476608 DOI: 10.3390/ijms251910514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/25/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Sirtuin 5 (SIRT5) plays an important role in the maintenance of lipid metabolism and in white adipose tissue browning. In this study, we established a mouse model for diet-induced obesity and the browning of white fat; combined with gene expression intervention, transcriptome sequencing, and cell molecular biology methods, the regulation and molecular mechanisms of SIRT5 on fat deposition and beige fat formation were studied. The results showed that the loss of SIRT5 in obese mice exacerbated white adipose tissue deposition and metabolic inflexibility. Furthermore, the deletion of SIRT5 in a white-fat-browning mouse increased the succinylation of uncoupling protein 1 (UCP1), resulting in a loss of the beiging capacity of the subcutaneous white adipose tissue and impaired cold tolerance. Mechanistically, the inhibition of SIRT5 results in impaired CCAAT/enhancer binding protein beta (C/EBPβ) expression in brown adipocytes, which in turn reduces the UCP1 transcriptional pathway. Thus, the transcription of UCP1 mediated by the SIRT5-C/EBPβ axis is critical in regulating energy balance and obesity-related metabolism.
Collapse
Affiliation(s)
| | | | | | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China; (X.Z.); (L.D.); (S.W.)
| |
Collapse
|
19
|
Wang T, Sharma AK, Wu C, Maushart CI, Ghosh A, Yang W, Stefanicka P, Kovanicova Z, Ukropec J, Zhang J, Arnold M, Klug M, De Bock K, Schneider U, Popescu C, Zheng B, Ding L, Long F, Dewal RS, Moser C, Sun W, Dong H, Takes M, Suelberg D, Mameghani A, Nocito A, Zech CJ, Chirindel A, Wild D, Burger IA, Schön MR, Dietrich A, Gao M, Heine M, Sun Y, Vargas-Castillo A, Søberg S, Scheele C, Balaz M, Blüher M, Betz MJ, Spiegelman BM, Wolfrum C. Single-nucleus transcriptomics identifies separate classes of UCP1 and futile cycle adipocytes. Cell Metab 2024; 36:2130-2145.e7. [PMID: 39084216 DOI: 10.1016/j.cmet.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/22/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024]
Abstract
Adipose tissue can recruit catabolic adipocytes that utilize chemical energy to dissipate heat. This process occurs either by uncoupled respiration through uncoupling protein 1 (UCP1) or by utilizing ATP-dependent futile cycles (FCs). However, it remains unclear how these pathways coexist since both processes rely on the mitochondrial membrane potential. Utilizing single-nucleus RNA sequencing to deconvolute the heterogeneity of subcutaneous adipose tissue in mice and humans, we identify at least 2 distinct subpopulations of beige adipocytes: FC-adipocytes and UCP1-beige adipocytes. Importantly, we demonstrate that the FC-adipocyte subpopulation is highly metabolically active and utilizes FCs to dissipate energy, thus contributing to thermogenesis independent of Ucp1. Furthermore, FC-adipocytes are important drivers of systemic energy homeostasis and linked to glucose metabolism and obesity resistance in humans. Taken together, our findings identify a noncanonical thermogenic adipocyte subpopulation, which could be an important regulator of energy homeostasis in mammals.
Collapse
Affiliation(s)
- Tongtong Wang
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Chunyan Wu
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Claudia Irene Maushart
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital of Basel and University of Basel, Basel, Switzerland
| | - Adhideb Ghosh
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Wu Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Patrik Stefanicka
- Department of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine and University Hospital, Comenius University in Bratislava, Bratislava, Slovakia
| | - Zuzana Kovanicova
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jozef Ukropec
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia
| | - Jing Zhang
- Laboratory of Exercise and Health, Health Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Myrtha Arnold
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Manuel Klug
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Katrien De Bock
- Laboratory of Exercise and Health, Health Institute of Human Movement Sciences and Sport, Department of Health Sciences and Technology, ETH Zurich, Schwerzenbach, Switzerland
| | - Ulrich Schneider
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Cristina Popescu
- Department of Nuclear Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Bo Zheng
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Lianggong Ding
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Fen Long
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Revati Sumukh Dewal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Caroline Moser
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Wenfei Sun
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Hua Dong
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland
| | - Martin Takes
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Dominique Suelberg
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Alexander Mameghani
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Antonio Nocito
- Department of Surgery, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland
| | - Christoph Johannes Zech
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Alin Chirindel
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Damian Wild
- Department of Radiology and Nuclear Medicine, University Hospital of Basel, Basel, Switzerland
| | - Irene A Burger
- Department of Nuclear Medicine, Cantonal Hospital of Baden, Im Ergel 1, 5404 Baden, Switzerland; Department of Nuclear Medicine, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Michael R Schön
- Städtisches Klinikum Karlsruhe, Clinic of Visceral Surgery, Karlsruhe, Germany
| | - Arne Dietrich
- Clinic for Visceral, Transplant and Thoracic and Vascular Surgery, University Hospital Leipzig, Liebigstrasse 20, 04103 Leipzig, Germany
| | - Min Gao
- Department of Pharmacy, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Markus Heine
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Yizhi Sun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Ariana Vargas-Castillo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Susanna Søberg
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Center of Inflammation and Metabolism and the Center for Physical Activity Research, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Camilla Scheele
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Center of Inflammation and Metabolism and the Center for Physical Activity Research, Rigshospitalet, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Miroslav Balaz
- Institute of Experimental Endocrinology, Biomedical Research Center at the Slovak Academy of Sciences, Bratislava, Slovakia; Department of Animal Physiology and Ethology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Germany & Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital, Leipzig, Germany.
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital of Basel and University of Basel, Basel, Switzerland.
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA; Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, Department of Health Sciences and Technology ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
20
|
Bardova K, Janovska P, Vavrova A, Kopecky J, Zouhar P. Adaptive Induction of Nonshivering Thermogenesis in Muscle Rather Than Brown Fat Could Counteract Obesity. Physiol Res 2024; 73:S279-S294. [PMID: 38752772 PMCID: PMC11412341 DOI: 10.33549/physiolres.935361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024] Open
Abstract
Warm-blooded animals such as birds and mammals are able to protect stable body temperature due to various thermogenic mechanisms. These processes can be facultative (occurring only under specific conditions, such as acute cold) and adaptive (adjusting their capacity according to long-term needs). They can represent a substantial part of overall energy expenditure and, therefore, affect energy balance. Classical mechanisms of facultative thermogenesis include shivering of skeletal muscles and (in mammals) non-shivering thermogenesis (NST) in brown adipose tissue (BAT), which depends on uncoupling protein 1 (UCP1). Existence of several alternative thermogenic mechanisms has been suggested. However, their relative contribution to overall heat production and the extent to which they are adaptive and facultative still needs to be better defined. Here we focus on comparison of NST in BAT with thermogenesis in skeletal muscles, including shivering and NST. We present indications that muscle NST may be adaptive but not facultative, unlike UCP1-dependent NST. Due to its slow regulation and low energy efficiency, reflecting in part the anatomical location, induction of muscle NST may counteract development of obesity more effectively than UCP1-dependent thermogenesis in BAT.
Collapse
Affiliation(s)
- K Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Prague 4, Czech Republic. or
| | | | | | | | | |
Collapse
|
21
|
Lee AH, Orliaguet L, Youm YH, Maeda R, Dlugos T, Lei Y, Coman D, Shchukina I, Andhey S, Smith SR, Ravussin E, Stadler K, Hyder F, Artyomov MN, Sugiura Y, Dixit VD. Cysteine depletion triggers adipose tissue thermogenesis and weight-loss. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.06.606880. [PMID: 39149397 PMCID: PMC11326254 DOI: 10.1101/2024.08.06.606880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Dietary interventions such as caloric restriction (CR)1 and methionine restriction2 that prolong lifespan induce the 'browning' of white adipose tissue (WAT), an adaptive metabolic response that increases heat production to maintain health3,4. However, how diet influences adipose browning and metabolic health is unclear. Here, we identified that weight-loss induced by CR in humans5 reduces cysteine concentration in WAT suggesting depletion of this amino-acid may be involved in metabolic benefits of CR. To investigate the role of cysteine on organismal metabolism, we created a cysteine-deficiency mouse model in which dietary cysteine was eliminated and cystathionine γ-lyase (CTH)6, the enzyme that synthesizes cysteine was conditionally deleted. Using this animal model, we found that systemic cysteine-depletion causes drastic weight-loss with increased fat utilization and browning of adipose tissue. The restoration of dietary cysteine in cysteine-deficient mice rescued weight loss together with reversal of adipose browning and increased food-intake in an on-demand fashion. Mechanistically, cysteine deficiency induced browning and weight loss is dependent on sympathetic nervous system derived noradrenaline signaling via β3-adrenergic-receptors and does not require UCP1. Therapeutically, in high-fat diet fed obese mice, one week of cysteine-deficiency caused 30% weight-loss and reversed inflammation. These findings thus establish that cysteine is essential for organismal metabolism as removal of cysteine in the host triggers adipose browning and rapid weight loss.
Collapse
Affiliation(s)
- Aileen H. Lee
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Lucie Orliaguet
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yun-Hee Youm
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | | | - Tamara Dlugos
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Yuanjiu Lei
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Daniel Coman
- Department of Radiology and Biomedical Imaging, School of Engineering and Applied Science, Yale University
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University
| | - Irina Shchukina
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Sairam Andhey
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth, Orlando, FL, USA
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | | | - Fahmeed Hyder
- Department of Radiology and Biomedical Imaging, School of Engineering and Applied Science, Yale University
- Department of Biomedical Engineering, School of Engineering and Applied Science, Yale University
| | - Maxim N. Artyomov
- Department of Pathology and Immunology Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | - Vishwa Deep Dixit
- Department of Pathology, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT 06520, USA
- Yale Center for Research on Aging, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
22
|
Yuan B, Doxsey W, Tok Ö, Kwon YY, Inouye KE, Hotamışlıgil GS, Hui S. An Organism-Level Quantitative Flux Model of Energy Metabolism in Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.11.579776. [PMID: 38405872 PMCID: PMC10888810 DOI: 10.1101/2024.02.11.579776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Mammalian tissues feed on nutrients in the blood circulation. At the organism-level, mammalian energy metabolism comprises of oxidation, storage, interconverting, and releasing of circulating nutrients. Though much is known about the individual processes and nutrients, a holistic and quantitative model describing these processes for all major circulating nutrients is lacking. Here, by integrating isotope tracer infusion, mass spectrometry, and isotope gas analyzer measurement, we developed a framework to systematically quantify fluxes through these metabolic processes for 10 major circulating energy nutrients in mice, resulting in an organism-level quantitative flux model of energy metabolism. This model revealed in wildtype mice that circulating nutrients have more dominant metabolic cycling fluxes than their oxidation fluxes, with distinct partition between cycling and oxidation flux for individual circulating nutrients. Applications of this framework in obese mouse models showed on a per animal basis extensive elevation of metabolic cycling fluxes in ob/ob mice, but not in diet-induced obese mice. Thus, our framework describes quantitatively the functioning of energy metabolism at the organism-level, valuable for revealing new features of energy metabolism in physiological and disease conditions. Highlights A flux model of energy metabolism integrating 13 C labeling of metabolites and CO 2 Circulating nutrients have characteristic partition between oxidation and storageCirculating nutrients' total cycling flux outweighs their total oxidation fluxCycling fluxes are extensively elevated in ob/ob but not in diet-induced obese mice.
Collapse
|
23
|
Natarajan D, Plakkot B, Tiwari K, Ekambaram S, Wang W, Rudolph M, Mohammad MA, Chacko SK, Subramanian M, Tarantini S, Yabluchanskiy A, Ungvari Z, Csiszar A, Balasubramanian P. The metabolic benefits of thermogenic stimulation are preserved in aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601572. [PMID: 39005396 PMCID: PMC11244901 DOI: 10.1101/2024.07.01.601572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Adipose thermogenesis has been actively investigated as a therapeutic target for improving metabolic dysfunction in obesity. However, its applicability to middle-aged and older populations, which bear the highest obesity prevalence in the US (approximately 40%), remains uncertain due to age-related decline in thermogenic responses. In this study, we investigated the effects of chronic thermogenic stimulation using the β3-adrenergic (AR) agonist CL316,243 (CL) on systemic metabolism and adipose function in aged (18-month-old) C57BL/6JN mice. Sustained β3-AR treatment resulted in reduced fat mass, increased energy expenditure, increased fatty acid oxidation and mitochondrial activity in adipose depots, improved glucose homeostasis, and a favorable adipokine profile. At the cellular level, CL treatment increased uncoupling protein 1 (UCP1)-dependent thermogenesis in brown adipose tissue (BAT). However, in white adipose tissue (WAT) depots, CL treatment increased glycerol and lipid de novo lipogenesis (DNL) and turnover suggesting the activation of the futile substrate cycle of lipolysis and reesterification in a UCP1-independent manner. Increased lipid turnover was also associated with the simultaneous upregulation of proteins involved in glycerol metabolism, fatty acid oxidation, and reesterification in WAT. Further, a dose-dependent impact of CL treatment on inflammation was observed, particularly in subcutaneous WAT, suggesting a potential mismatch between fatty acid supply and oxidation. These findings indicate that chronic β3-AR stimulation activates distinct cellular mechanisms that increase energy expenditure in BAT and WAT to improve systemic metabolism in aged mice. Our study provides foundational evidence for targeting adipose thermogenesis to improve age-related metabolic dysfunction.
Collapse
|
24
|
Su Q, Raza SHA, Gao Z, Zhang F, Wu Z, Ji Q, He T, Aloufi BH, El-Mansi AA, Eldesoqui M, Sabir DK, Gui L. Profiling and functional analysis of circular RNAs in yaks intramuscular fat. J Anim Physiol Anim Nutr (Berl) 2024; 108:1016-1027. [PMID: 38432684 DOI: 10.1111/jpn.13947] [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: 11/10/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
Circular RNAs (circRNAs) are a new class of endogenous RNA regulating gene expression. However, the regulatory mechanisms of lipid metabolism in yaks involved in circRNAs remain poorly understood. The IMF plays a crucial role in the quality of yak meat, to greatly improve the meat quality. In this study, the fatty acid profiles of yak IMF were determined and circRNAs were sequenced. The results showed that the total of polyunsaturated fatty acid (PUFA) content of adult yak muscle was significantly higher than that in yak calves (p < 0.05). A total of 29,021 circRNAs were identified in IMF tissue, notably, 99 differentially expressed (DE) circRNAs were identified, to be associated with fat deposition, the most significant of which were circ_12686, circ_6918, circ_3582, ci_106 and ci_123 (A circRNA composed of exons is labelled 'circRNA' and a circRNA composed of introns is labelled 'ciRNA'). KEGG pathway enrichment analysis showed that the differential circRNAs were enriched in four pathways associated with fat deposition (e.g., the peroxisome proliferator-activated receptor signalling, fatty acid degradation, sphingolipid metabolism and sphingolipid signalling pathways). We also constructed co-expression networks of DE circRNA-miRNA using high-throughput sequencing in IMF deposition, from which revealed that ci_106 target binding of bta-miR-130b, bta-miR-148a, bta-miR-15a, bta-miR-34a, bta-miR-130a, bta-miR-17-5p and ci_123 target binding of bta-miR-150 were involved in adipogenesis. The study revealed the role of the circRNAs in the IMF deposition in yak and its influence on meat quality the findings demonstrated the circRNA differences in the development of IMF with the increase of age, thus providing a theoretical basis for further research on the molecular mechanism of IMF deposition in yaks.
Collapse
Affiliation(s)
- Quyangangmao Su
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - Sayed Haidar Abbas Raza
- Guangdong Provincial Key Laboratory of Food Quality and Safet, Nation-Local Joint Engineering Research Center for Machining and Safety of Livestock and Poultry Products, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan, China
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Zhanhong Gao
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - Fengshuo Zhang
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - ZhenLing Wu
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - QiuRong Ji
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - TingLi He
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| | - Bandar Hamad Aloufi
- Biology Department, Faculty of Science, University of Ha'il, Ha'il, Saudi Arabia
| | - Ahmed A El-Mansi
- Biology Department, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Mamdouh Eldesoqui
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Diriyah, Riyadh, Saudi Arabia
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Deema Kamal Sabir
- Department of Medical Surgical Nursing, College of Nursing, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Linsheng Gui
- College of Agriculture and Animal Husbandry, Qinghai University, Xining, People's Republic of China
| |
Collapse
|
25
|
Sahin C, Melanson JR, Le Billan F, Magomedova L, Ferreira TAM, Oliveira AS, Pollock-Tahari E, Saikali MF, Cash SB, Woo M, Romeiro LAS, Cummins CL. A novel fatty acid mimetic with pan-PPAR partial agonist activity inhibits diet-induced obesity and metabolic dysfunction-associated steatotic liver disease. Mol Metab 2024; 85:101958. [PMID: 38763495 PMCID: PMC11170206 DOI: 10.1016/j.molmet.2024.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/21/2024] Open
Abstract
OBJECTIVE The prevalence of metabolic diseases is increasing globally at an alarming rate; thus, it is essential that effective, accessible, low-cost therapeutics are developed. Peroxisome proliferator-activated receptors (PPARs) are transcription factors that tightly regulate glucose homeostasis and lipid metabolism and are important drug targets for the treatment of type 2 diabetes and dyslipidemia. We previously identified LDT409, a fatty acid-like compound derived from cashew nut shell liquid, as a novel pan-active PPARα/γ/δ compound. Herein, we aimed to assess the efficacy of LDT409 in vivo and investigate the molecular mechanisms governing the actions of the fatty acid mimetic LDT409 in diet-induced obese mice. METHODS C57Bl/6 mice (6-11-month-old) were fed a chow or high fat diet (HFD) for 4 weeks; mice thereafter received once daily intraperitoneal injections of vehicle, 10 mg/kg Rosiglitazone, 40 mg/kg WY14643, or 40 mg/kg LDT409 for 18 days while continuing the HFD. During treatments, body weight, food intake, glucose and insulin tolerance, energy expenditure, and intestinal lipid absorption were measured. On day 18 of treatment, tissues and plasma were collected for histological, molecular, and biochemical analysis. RESULTS We found that treatment with LDT409 was effective at reversing HFD-induced obesity and associated metabolic abnormalities in mice. LDT409 lowered food intake and hyperlipidemia, while improving insulin tolerance. Despite being a substrate of both PPARα and PPARγ, LDT409 was crucial for promoting hepatic fatty acid oxidation and reducing hepatic steatosis in HFD-fed mice. We also highlighted a role for LDT409 in white and brown adipocytes in vitro and in vivo where it decreased fat accumulation, increased lipolysis, induced browning of WAT, and upregulated thermogenic gene Ucp1. Remarkably, LDT409 reversed HFD-induced weight gain back to chow-fed control levels. We determined that the LDT409-induced weight-loss was associated with a combination of increased energy expenditure (detectable before weight loss was apparent), decreased food intake, increased systemic fat utilization, and increased fecal lipid excretion in HFD-fed mice. CONCLUSIONS Collectively, LDT409 represents a fatty acid mimetic that generates a uniquely favorable metabolic response for the treatment of multiple abnormalities including obesity, dyslipidemia, metabolic dysfunction-associated steatotic liver disease, and diabetes. LDT409 is derived from a highly abundant natural product-based starting material and its development could be pursued as a therapeutic solution to the global metabolic health crisis.
Collapse
Affiliation(s)
- Cigdem Sahin
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Jenna-Rose Melanson
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Florian Le Billan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Lilia Magomedova
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Thais A M Ferreira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Andressa S Oliveira
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Evan Pollock-Tahari
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Michael F Saikali
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Sarah B Cash
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Minna Woo
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, M5G 2C4, Canada; Banting and Best Diabetes Centre, Toronto, ON, M5G 2C4, Canada
| | - Luiz A S Romeiro
- Department of Pharmacy, Faculty of Health Sciences, University of Brasilia, Brasilia, DF 71910-900, Brazil
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada; Banting and Best Diabetes Centre, Toronto, ON, M5G 2C4, Canada.
| |
Collapse
|
26
|
Sharma AK, Khandelwal R, Wolfrum C. Futile cycles: Emerging utility from apparent futility. Cell Metab 2024; 36:1184-1203. [PMID: 38565147 DOI: 10.1016/j.cmet.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/15/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Futile cycles are biological phenomena where two opposing biochemical reactions run simultaneously, resulting in a net energy loss without appreciable productivity. Such a state was presumed to be a biological aberration and thus deemed an energy-wasting "futile" cycle. However, multiple pieces of evidence suggest that biological utilities emerge from futile cycles. A few established functions of futile cycles are to control metabolic sensitivity, modulate energy homeostasis, and drive adaptive thermogenesis. Yet, the physiological regulation, implication, and pathological relevance of most futile cycles remain poorly studied. In this review, we highlight the abundance and versatility of futile cycles and propose a classification scheme. We further discuss the energetic implications of various futile cycles and their impact on basal metabolic rate, their bona fide and tentative pathophysiological implications, and putative drug interactions.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
27
|
Carpentier AC, Blondin DP. Is stimulation of browning of human adipose tissue a relevant therapeutic target? ANNALES D'ENDOCRINOLOGIE 2024; 85:184-189. [PMID: 38871497 DOI: 10.1016/j.ando.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Brown adipose tissue (BAT) and beige adipose tissues are important contributors to cold-induced whole body thermogenesis in rodents. The documentation in humans of cold- and ß-adrenergic receptor agonist-stimulated BAT glucose uptake using positron emission tomography (PET) and of a decrease of this response in individuals with cardiometabolic disorders led to the suggestion that BAT/beige adipose tissues could be relevant targets for prevention and treatment of these conditions. In this brief review, we will critically assess this question by first describing the basic rationale for this affirmation, second by examining the evidence in human studies, and third by discussing the possible means to activate the thermogenic response of these tissues in humans.
Collapse
Affiliation(s)
- André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Sherbrooke, Québec, Canada.
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
28
|
Ma L, Gilani A, Rubio-Navarro A, Cortada E, Li A, Reilly SM, Tang L, Lo JC. Adipsin and adipocyte-derived C3aR1 regulate thermogenic fat in a sex-dependent fashion. JCI Insight 2024; 9:e178925. [PMID: 38713526 PMCID: PMC11382875 DOI: 10.1172/jci.insight.178925] [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: 12/27/2023] [Accepted: 04/26/2024] [Indexed: 05/09/2024] Open
Abstract
Thermogenesis in beige/brown adipose tissues can be leveraged to combat metabolic disorders such as type 2 diabetes and obesity. The complement system plays pleiotropic roles in metabolic homeostasis and organismal energy balance with canonical effects on immune cells and noncanonical effects on nonimmune cells. The adipsin/C3a/C3a receptor 1 (C3aR1) pathway stimulates insulin secretion and sustains pancreatic β cell mass. However, its role in adipose thermogenesis has not been defined. Here, we show that male Adipsin/Cfd-knockout mice exhibited increased energy expenditure and white adipose tissue (WAT) browning. In addition, male adipocyte-specific C3aR1-knockout mice exhibited enhanced WAT thermogenesis and increased respiration. In stark contrast, female adipocyte-specific C3aR1-knockout mice displayed decreased brown fat thermogenesis and were cold intolerant. Female mice expressed lower levels of Adipsin in thermogenic adipocytes and adipose tissues than males. C3aR1 was also lower in female subcutaneous adipose tissue than in males. Collectively, these results reveal sexual dimorphism in the adipsin/C3a/C3aR1 axis in regulating adipose thermogenesis and defense against cold stress. Our findings establish a potentially new role of the alternative complement pathway in adaptive thermogenesis and highlight sex-specific considerations in potential therapeutic targets for metabolic diseases.
Collapse
Affiliation(s)
- Lunkun Ma
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Ankit Gilani
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Alfonso Rubio-Navarro
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Eric Cortada
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Ang Li
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| | - Shannon M Reilly
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - James C Lo
- Division of Cardiology, Department of Medicine
- Weill Center for Metabolic Health; and
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
29
|
Mota INR, Satari S, Marques IS, Santos JMO, Medeiros R. Adipose tissue rearrangement in cancer cachexia: The involvement of β3-adrenergic receptor associated pathways. Biochim Biophys Acta Rev Cancer 2024; 1879:189103. [PMID: 38679401 DOI: 10.1016/j.bbcan.2024.189103] [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: 10/17/2023] [Revised: 04/08/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
Cancer-associated cachexia (CAC) is a complex multiple organ syndrome that significantly contributes to reduced quality of life and increased mortality among many cancer patients. Its multifactorial nature makes its early diagnosis and effective therapeutic interventions challenging. Adipose tissue is particularly impacted by cachexia, typically through increased lipolysis, browning and thermogenesis, mainly at the onset of the disease. These processes lead to depletion of fat mass and contribute to the dysfunction of other organs. The β-adrenergic signalling pathways are classical players in the regulation of adipose tissue metabolism. They are activated upon sympathetic stimulation inducing lipolysis, browning and thermogenesis, therefore contributing to energy expenditure. Despite accumulating evidence suggesting that β3-adrenergic receptor stimulation may be crucial to the adipose tissue remodelling during cachexia, the literature remains controversial. Moreover, there is limited knowledge regarding sexual dimorphism of adipose tissue in the context of cachexia. This review paper aims to present the current knowledge regarding adipose tissue wasting during CAC, with a specific focus on the role of the β3-adrenergic receptor, placing it as a potential therapeutic target against cachexia.
Collapse
Affiliation(s)
- Inês N R Mota
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal.
| | - Setareh Satari
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal.
| | - Inês Soares Marques
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Faculty of Sciences, University of Porto (FCUP), 4169-007 Porto, Portugal.
| | - Joana M O Santos
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Research Department of the Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), 4200-172 Porto, Portugal.
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP)/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center Raquel Seruca (Porto.CCC), 4200-072 Porto, Portugal; Research Department of the Portuguese League Against Cancer - Regional Nucleus of the North (Liga Portuguesa Contra o Cancro - Núcleo Regional do Norte), 4200-172 Porto, Portugal; Virology Service, Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal; Biomedical Research Center (CEBIMED), Faculty of Health Sciences of the Fernando Pessoa University, 4249-004 Porto, Portugal.
| |
Collapse
|
30
|
Sharma AK, Khandelwal R, Wolfrum C. Futile lipid cycling: from biochemistry to physiology. Nat Metab 2024; 6:808-824. [PMID: 38459186 DOI: 10.1038/s42255-024-01003-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/02/2024] [Indexed: 03/10/2024]
Abstract
In the healthy state, the fat stored in our body isn't just inert. Rather, it is dynamically mobilized to maintain an adequate concentration of fatty acids (FAs) in our bloodstream. Our body tends to produce excess FAs to ensure that the FA availability is not limiting. The surplus FAs are actively re-esterified into glycerides, initiating a cycle of breakdown and resynthesis of glycerides. This cycle consumes energy without generating a new product and is commonly referred to as the 'futile lipid cycle' or the glyceride/FA cycle. Contrary to the notion that it's a wasteful process, it turns out this cycle is crucial for systemic metabolic homeostasis. It acts as a control point in intra-adipocyte and inter-organ cross-talk, a metabolic rheostat, an energy sensor and a lipid diversifying mechanism. In this Review, we discuss the metabolic regulation and physiological implications of the glyceride/FA cycle and its mechanistic underpinnings.
Collapse
Affiliation(s)
- Anand Kumar Sharma
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Radhika Khandelwal
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland
| | - Christian Wolfrum
- Laboratory of Translational Nutrition Biology, Institute of Food, Nutrition and Health, ETH Zurich, Schwerzenbach, Switzerland.
| |
Collapse
|
31
|
Zhang Y, Zhang B, Sun X. The molecular mechanism of macrophage-adipocyte crosstalk in maintaining energy homeostasis. Front Immunol 2024; 15:1378202. [PMID: 38650945 PMCID: PMC11033412 DOI: 10.3389/fimmu.2024.1378202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/25/2024] [Indexed: 04/25/2024] Open
Abstract
Interactions between macrophages and adipocytes in adipose tissue are critical for the regulation of energy metabolism and obesity. Macrophage polarization induced by cold or other stimulations can drive metabolic reprogramming of adipocytes, browning, and thermogenesis. Accordingly, investigating the roles of macrophages and adipocytes in the maintenance of energy homeostasis is critical for the development of novel therapeutic approaches specifically targeting macrophages in metabolic disorders such as obesity. Current review outlines macrophage polarization not only regulates the release of central nervous system and inflammatory factors, but controls mitochondrial function, and other factor that induce metabolic reprogramming of adipocytes and maintain energy homeostasis. We also emphasized on how the adipocytes conversely motivate the polarization of macrophage. Exploring the interactions between adipocytes and macrophages may provide new therapeutic strategies for the management of obesity-related metabolic diseases.
Collapse
Affiliation(s)
- Yudie Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Bin Zhang
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, China
- Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Beijing, China
- Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Beijing, China
- Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glyeolipid Metabolism Disorder Disease, State Administration of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Yu X, Benitez G, Wei PT, Krylova SV, Song Z, Liu L, Zhang M, Xiaoli AM, Wei H, Chen F, Sidoli S, Yang F, Shinoda K, Pessin JE, Feng D. Involution of brown adipose tissue through a Syntaxin 4 dependent pyroptosis pathway. Nat Commun 2024; 15:2856. [PMID: 38565851 PMCID: PMC10987578 DOI: 10.1038/s41467-024-46944-y] [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: 06/15/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Aging, chronic high-fat diet feeding, or housing at thermoneutrality induces brown adipose tissue (BAT) involution, a process characterized by reduction of BAT mass and function with increased lipid droplet size. Single nuclei RNA sequencing of aged mice identifies a specific brown adipocyte population of Ucp1-low cells that are pyroptotic and display a reduction in the longevity gene syntaxin 4 (Stx4a). Similar to aged brown adipocytes, Ucp1-STX4KO mice display loss of brown adipose tissue mass and thermogenic dysfunction concomitant with increased pyroptosis. Restoration of STX4 expression or suppression of pyroptosis activation protects against the decline in both mass and thermogenic activity in the aged and Ucp1-STX4KO mice. Mechanistically, STX4 deficiency reduces oxidative phosphorylation, glucose uptake, and glycolysis leading to reduced ATP levels, a known triggering signal for pyroptosis. Together, these data demonstrate an understanding of rapid brown adipocyte involution and that physiologic aging and thermogenic dysfunction result from pyroptotic signaling activation.
Collapse
Affiliation(s)
- Xiaofan Yu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Gabrielle Benitez
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Peter Tszki Wei
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, 14853, USA
| | - Sofia V Krylova
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ziyi Song
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, 530004, China
| | - Li Liu
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Meifan Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, 08854, USA
| | - Alus M Xiaoli
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Henna Wei
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Fenfen Chen
- Department of Animal Science, College of Life Science, Southwest Forestry University, Kunming, Yunnan, 650244, China
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Fajun Yang
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Kosaku Shinoda
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jeffrey E Pessin
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Daorong Feng
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
- Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| |
Collapse
|
33
|
Morocho-Jaramillo PA, Kotlar-Goldaper I, Zakarauskas-Seth BI, Purfürst B, Filosa A, Sawamiphak S. The zebrafish heart harbors a thermogenic beige fat depot analog of human epicardial adipose tissue. Cell Rep 2024; 43:113955. [PMID: 38507414 DOI: 10.1016/j.celrep.2024.113955] [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: 04/14/2023] [Revised: 01/25/2024] [Accepted: 02/28/2024] [Indexed: 03/22/2024] Open
Abstract
Epicardial adipose tissue (eAT) is a metabolically active fat depot that has been associated with a wide array of cardiac homeostatic functions and cardiometabolic diseases. A full understanding of its diverse physiological and pathological roles is hindered by the dearth of animal models. Here, we show, in the heart of an ectothermic teleost, the zebrafish, the existence of a fat depot localized underneath the epicardium, originating from the epicardium and exhibiting the molecular signature of beige adipocytes. Moreover, a subset of adipocytes within this cardiac fat tissue exhibits primitive thermogenic potential. Transcriptomic profiling and cross-species analysis revealed elevated glycolytic and cardiac homeostatic gene expression with downregulated obesity and inflammatory hallmarks in the teleost eAT compared to that of lean aged humans. Our findings unveil epicardium-derived beige fat in the heart of an ectotherm considered to possess solely white adipocytes for energy storage and identify pathways that may underlie age-driven remodeling of human eAT.
Collapse
Affiliation(s)
- Paul-Andres Morocho-Jaramillo
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Ilan Kotlar-Goldaper
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bhakti I Zakarauskas-Seth
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Bettina Purfürst
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Alessandro Filosa
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany
| | - Suphansa Sawamiphak
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str. 10, 13125 Berlin, Germany.
| |
Collapse
|
34
|
Rahbani JF, Bunk J, Lagarde D, Samborska B, Roesler A, Xiao H, Shaw A, Kaiser Z, Braun JL, Geromella MS, Fajardo VA, Koza RA, Kazak L. Parallel control of cold-triggered adipocyte thermogenesis by UCP1 and CKB. Cell Metab 2024; 36:526-540.e7. [PMID: 38272036 DOI: 10.1016/j.cmet.2024.01.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 01/27/2024]
Abstract
That uncoupling protein 1 (UCP1) is the sole mediator of adipocyte thermogenesis is a conventional viewpoint that has primarily been inferred from the attenuation of the thermogenic output of mice genetically lacking Ucp1 from birth (germline Ucp1-/-). However, germline Ucp1-/- mice harbor secondary changes within brown adipose tissue. To mitigate these potentially confounding ancillary changes, we constructed mice with inducible adipocyte-selective Ucp1 disruption. We find that, although germline Ucp1-/- mice succumb to cold-induced hypothermia with complete penetrance, most mice with the inducible deletion of Ucp1 maintain homeothermy in the cold. However, inducible adipocyte-selective co-deletion of Ucp1 and creatine kinase b (Ckb, an effector of UCP1-independent thermogenesis) exacerbates cold intolerance. Following UCP1 deletion or UCP1/CKB co-deletion from mature adipocytes, moderate cold exposure triggers the regeneration of mature brown adipocytes that coordinately restore UCP1 and CKB expression. Our findings suggest that thermogenic adipocytes utilize non-paralogous protein redundancy-through UCP1 and CKB-to promote cold-induced energy dissipation.
Collapse
Affiliation(s)
- Janane F Rahbani
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Jakub Bunk
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Damien Lagarde
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Bozena Samborska
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Anna Roesler
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Haopeng Xiao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Abhirup Shaw
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
| | - Zafir Kaiser
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Mia S Geromella
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Robert A Koza
- MaineHealth Institute for Research, Scarborough, ME 04074, USA
| | - Lawrence Kazak
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada.
| |
Collapse
|
35
|
Stanic S, Bardova K, Janovska P, Rossmeisl M, Kopecky J, Zouhar P. Prolonged FGF21 treatment increases energy expenditure and induces weight loss in obese mice independently of UCP1 and adrenergic signaling. Biochem Pharmacol 2024; 221:116042. [PMID: 38325495 DOI: 10.1016/j.bcp.2024.116042] [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: 11/15/2023] [Revised: 12/22/2023] [Accepted: 02/01/2024] [Indexed: 02/09/2024]
Abstract
Fibroblast growth factor 21 (FGF21) reduces body weight, which was attributed to induced energy expenditure (EE). Conflicting data have been published on the role of uncoupling protein 1 (UCP1) in this effect. Therefore, we aimed to revisit the thermoregulatory effects of FGF21 and their implications for body weight regulation. We found that an 8-day treatment with FGF21 lowers body weight to similar extent in both wildtype (WT) and UCP1-deficient (KO) mice fed high-fat diet. In WT mice, this effect is solely due to increased EE, associated with a strong activation of UCP1 and with excess heat dissipated through the tail. This thermogenesis takes place in the interscapular region and can be attenuated by a β-adrenergic inhibitor propranolol. In KO mice, FGF21-induced weight loss correlates with a modest increase in EE, which is independent of adrenergic signaling, and with a reduced energy intake. Interestingly, the gene expression profile of interscapular brown adipose tissue (but not subcutaneous white adipose tissue) of KO mice is massively affected by FGF21, as shown by increased expression of genes encoding triacylglycerol/free fatty acid cycle enzymes. Thus, FGF21 elicits central thermogenic and pyretic effects followed by a concomitant increase in EE and body temperature, respectively. The associated weight loss is strongly dependent on UCP1-based thermogenesis. However, in the absence of UCP1, alternative mechanisms of energy dissipation may contribute, possibly based on futile triacylglycerol/free fatty acid cycling in brown adipose tissue and reduced food intake.
Collapse
Affiliation(s)
- Sara Stanic
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic; Faculty of Science, Charles University in Prague, Vinicna 7, Prague 128 44, Czech Republic
| | - Kristina Bardova
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| | - Petra Janovska
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| | - Martin Rossmeisl
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| | - Jan Kopecky
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic
| | - Petr Zouhar
- Laboratory of Adipose Tissue Biology, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 142 00, Czech Republic.
| |
Collapse
|
36
|
Naren Q, Lindsund E, Bokhari MH, Pang W, Petrovic N. Differential responses to UCP1 ablation in classical brown versus beige fat, despite a parallel increase in sympathetic innervation. J Biol Chem 2024; 300:105760. [PMID: 38367663 PMCID: PMC10944106 DOI: 10.1016/j.jbc.2024.105760] [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: 08/13/2023] [Revised: 01/27/2024] [Accepted: 02/09/2024] [Indexed: 02/19/2024] Open
Abstract
In the cold, the absence of the mitochondrial uncoupling protein 1 (UCP1) results in hyper-recruitment of beige fat, but classical brown fat becomes atrophied. Here we examine possible mechanisms underlying this phenomenon. We confirm that in brown fat from UCP1-knockout (UCP1-KO) mice acclimated to the cold, the levels of mitochondrial respiratory chain proteins were diminished; however, in beige fat, the mitochondria seemed to be unaffected. The macrophages that accumulated massively not only in brown fat but also in beige fat of the UCP1-KO mice acclimated to cold did not express tyrosine hydroxylase, the norepinephrine transporter (NET) and monoamine oxidase-A (MAO-A). Consequently, they could not influence the tissues through the synthesis or degradation of norepinephrine. Unexpectedly, in the cold, both brown and beige adipocytes from UCP1-KO mice acquired an ability to express MAO-A. Adipose tissue norepinephrine was exclusively of sympathetic origin, and sympathetic innervation significantly increased in both tissues of UCP1-KO mice. Importantly, the magnitude of sympathetic innervation and the expression levels of genes induced by adrenergic stimulation were much higher in brown fat. Therefore, we conclude that no qualitative differences in innervation or macrophage character could explain the contrasting reactions of brown versus beige adipose tissues to UCP1-ablation. Instead, these contrasting responses may be explained by quantitative differences in sympathetic innervation: the beige adipose depot from the UCP1-KO mice responded to cold acclimation in a canonical manner and displayed enhanced recruitment, while the atrophy of brown fat lacking UCP1 may be seen as a consequence of supraphysiological adrenergic stimulation in this tissue.
Collapse
Affiliation(s)
- Qimuge Naren
- College of Animal Science and Technology, Northwest A&F University, Yangling, China; Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Erik Lindsund
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Muhammad Hamza Bokhari
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Weijun Pang
- College of Animal Science and Technology, Northwest A&F University, Yangling, China.
| | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden.
| |
Collapse
|
37
|
Michurina S, Agareva M, Zubkova E, Menshikov M, Stafeev I, Parfyonova Y. IL-4 activates the futile triacylglyceride cycle for glucose utilization in white adipocytes. Biochem J 2024; 481:329-344. [PMID: 38323641 DOI: 10.1042/bcj20230486] [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: 12/01/2023] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 02/08/2024]
Abstract
The development of cardiometabolic complications during obesity is strongly associated with chronic latent inflammation in hypertrophied adipose tissue (AT). IL-4 is an anti-inflammatory cytokine, playing a protective role against insulin resistance, glucose intolerance and weight gain. The positive effects of IL-4 are associated not only with the activation of anti-inflammatory immune cells in AT, but also with the modulation of adipocyte metabolism. IL-4 is known to activate lipolysis and glucose uptake in adipocytes, but the precise regulatory mechanisms and physiological significance of these processes remain unclear. In this study, we detail IL-4 effects on glucose and triacylglycerides (TAGs) metabolism and propose mechanisms of IL-4 metabolic action in adipocytes. We have shown that IL-4 activates glucose oxidation, lipid droplet (LD) fragmentation, lipolysis and thermogenesis in mature 3T3-L1 adipocytes. We found that lipolysis was not accompanied by fatty acids (FAs) release from adipocytes, suggesting FA re-esterification. Moreover, glucose oxidation and thermogenesis stimulation depended on adipocyte triglyceride lipase (ATGL) activity, but not the uncoupling protein (UCP1) expression. Based on these data, IL-4 may activate the futile TAG-FA cycle in adipocytes, which enhances the oxidative activity of cells and heat production. Thus, the positive effect of IL-4 on systemic metabolism can be the result of the activation of non-canonical thermogenic mechanism in AT, increasing TAG turnover and utilization of excessive glucose.
Collapse
Affiliation(s)
- Svetlana Michurina
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Margarita Agareva
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, 119991, Moscow, Russia
| | - Ekaterina Zubkova
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Mikhail Menshikov
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Iurii Stafeev
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
| | - Yelena Parfyonova
- Department of Angiogenesis, National Medical Research Centre for Cardiology named after academician E.I.Chazov, 121552, Moscow, Russia
- Faculty of Basic Medicine, Lomonosov Moscow State University, 119991, Moscow, Russia
| |
Collapse
|
38
|
Mota-Rojas D, Ghezzi MD, Hernández-Ávalos I, Domínguez-Oliva A, Casas-Alvarado A, Lendez PA, Ceriani MC, Wang D. Hypothalamic Neuromodulation of Hypothermia in Domestic Animals. Animals (Basel) 2024; 14:513. [PMID: 38338158 PMCID: PMC10854546 DOI: 10.3390/ani14030513] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/12/2024] Open
Abstract
When an organism detects decreases in their core body temperature, the hypothalamus, the main thermoregulatory center, triggers compensatory responses. These responses include vasomotor changes to prevent heat loss and physiological mechanisms (e.g., shivering and non-shivering thermogenesis) for heat production. Both types of changes require the participation of peripheral thermoreceptors, afferent signaling to the spinal cord and hypothalamus, and efferent pathways to motor and/or sympathetic neurons. The present review aims to analyze the scientific evidence of the hypothalamic control of hypothermia and the central and peripheral changes that are triggered in domestic animals.
Collapse
Affiliation(s)
- Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Marcelo Daniel Ghezzi
- Animal Welfare Area, Faculty of Veterinary Sciences (FCV), Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB, Tandil 7000, Buenos Aires, Argentina
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Biological Sciences Department, FESC, Universidad Nacional Autónoma de México, Cuautitlán 54714, Mexico
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Alejandro Casas-Alvarado
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana (UAM), Mexico City 04960, Mexico
| | - Pamela Anahí Lendez
- Anatomy Area, Faculty of Veterinary Sciences, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB/CISAPA, Tandil 7000, Buenos Aires, Argentina
| | - María Carolina Ceriani
- Anatomy Area, Faculty of Veterinary Sciences, Universidad Nacional del Centro de la Provincia de Buenos Aires (UNCPBA), GIB/CISAPA, Tandil 7000, Buenos Aires, Argentina
| | - Dehua Wang
- School of Life Sciences, Shandong University, Qingdao 266237, China
| |
Collapse
|
39
|
Li X, Feng L, Kuang Q, Wang X, Yang J, Niu X, Gao L, Huang L, Luo P, Li L. Microplastics cause hepatotoxicity in diabetic mice by disrupting glucolipid metabolism via PP2A/AMPK/HNF4A and promoting fibrosis via the Wnt/β-catenin pathway. ENVIRONMENTAL TOXICOLOGY 2024; 39:1018-1030. [PMID: 38064261 DOI: 10.1002/tox.24034] [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: 07/10/2023] [Revised: 10/24/2023] [Accepted: 10/31/2023] [Indexed: 01/09/2024]
Abstract
In recent years, microplastics (MPs) have gained significant attention as a persistent environmental pollutant resulting from the decomposition of plastics, leading to their accumulation in the human body. The liver, particularly of individuals with type 2 diabetes mellitus (T2DM), is known to be more susceptible to the adverse effects of environmental pollutants. Therefore, to investigate the potential impact of MPs on the liver of diabetic mice and elucidate the underlying toxicological mechanisms, we exposed db/db mice to 0.5 μm MPs for 3 months. Our results revealed that MPs exposure resulted in several harmful effects, including decreased body weight, disruption of liver structure and function, elevated blood glucose levels, impaired glucose tolerance, and increased glycogen accumulation in the hepatic tissue of the mice. Furthermore, MPs exposure was found to promote hepatic gluconeogenesis by perturbing the PP2A/AMPK/HNF4A signaling pathway. In addition, MPs disrupt redox balance, leading to oxidative damage in the liver. This exposure also disrupted hepatic lipid metabolism, stimulating lipid synthesis while inhibiting catabolism, ultimately resulting in the development of fatty liver. Moreover, MPs were found to induce liver fibrosis by activating the Wnt/β-catenin signaling pathway. Furthermore, MPs influenced adaptive thermogenesis in brown fat by modulating the expression of uncoupling protein 1 (UCP1) and genes associated with mitochondrial oxidative respiration thermogenesis in brown fat. In conclusion, our study demonstrates that MPs induce oxidative damage in the liver, disturb glucose and lipid metabolism, promote hepatic fibrosis, and influence adaptive thermogenesis in brown fat in diabetic mice. These findings underscore the potential adverse effects of MPs on liver health in individuals with T2DM and highlight the importance of further research in this area.
Collapse
Affiliation(s)
- Xinxin Li
- Department of Urology, Wuhan Third Hospital, Medical School of Wuhan University, Wuhan, China
| | - Lixiang Feng
- Department of Urology, Wuhan Third Hospital, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Qihui Kuang
- Department of Urology, Wuhan Third Hospital, Medical School of Wuhan University, Wuhan, China
| | - Xiong Wang
- Department of Pharmacy, Wuhan Third Hospital, Medical School of Wuhan University, Wuhan, China
| | - Jun Yang
- Department of Urology, Wuhan Third Hospital, Medical School of Wuhan University, Wuhan, China
| | - Xuan Niu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Likun Gao
- Department of Pathology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, China
| | - Lizhi Huang
- School of Civil Engineering, Wuhan University, Wuhan, China
| | - Pengcheng Luo
- Department of Urology, Wuhan Third Hospital, Medical School of Wuhan University, Wuhan, China
| | - Lili Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
40
|
El-Yazbi AF, Elrewiny MA, Habib HM, Eid AH, Elzahhar PA, Belal ASF. Thermogenic Modulation of Adipose Depots: A Perspective on Possible Therapeutic Intervention with Early Cardiorenal Complications of Metabolic Impairment. Mol Pharmacol 2023; 104:187-194. [PMID: 37567782 DOI: 10.1124/molpharm.123.000704] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular complications of diabetes and obesity remain a major cause for morbidity and mortality worldwide. Despite significant advances in the pharmacotherapy of metabolic disease, the available approaches do not prevent or slow the progression of complications. Moreover, a majority of patients present with significant vascular involvement at early stages of dysfunction prior to overt metabolic changes. The lack of disease-modifying therapies affects millions of patients globally, causing a massive economic burden due to these complications. Significantly, adipose tissue inflammation was implicated in the pathogenesis of metabolic syndrome, diabetes, and obesity. Specifically, perivascular adipose tissue (PVAT) and perirenal adipose tissue (PRAT) depots influence cardiovascular and renal structure and function. Accumulating evidence implicates localized PVAT/PRAT inflammation as the earliest response to metabolic impairment leading to cardiorenal dysfunction. Increased mitochondrial uncoupling protein 1 (UCP1) expression and function lead to PVAT/PRAT hypoxia and inflammation as well as vascular, cardiac, and renal dysfunction. As UCP1 function remains an undruggable target so far, modulation of the augmented UCP1-mediated PVAT/PRAT thermogenesis constitutes a lucrative target for drug development to mitigate early cardiorenal involvement. This can be achieved either by subtle targeted reduction in UCP-1 expression using innovative proteolysis activating chimeric molecules (PROTACs) or by supplementation with cyclocreatine phosphate, which augments the mitochondrial futile creatine cycling and thus decreases UCP1 activity, enhances the efficiency of oxygen use, and reduces hypoxia. Once developed, these molecules will be first-in-class therapeutic tools to directly interfere with and reverse the earliest pathology underlying cardiac, vascular, and renal dysfunction accompanying the early metabolic deterioration. SIGNIFICANCE STATEMENT: Adipose tissue dysfunction plays a major role in the pathogenesis of metabolic diseases and their complications. Although mitochondrial alterations are common in metabolic impairment, it was only recently shown that the early stages of metabolic challenge involve inflammatory changes in select adipose depots associated with increased uncoupling protein 1 thermogenesis and hypoxia. Manipulating this mode of thermogenesis can help mitigate the early inflammation and the consequent cardiorenal complications.
Collapse
Affiliation(s)
- Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Mohamed A Elrewiny
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Hosam M Habib
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ali H Eid
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Perihan A Elzahhar
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| | - Ahmed S F Belal
- Department of Pharmacology and Toxicology (A.F.E.-Y.) and Department of Pharmaceutical Chemistry (P.A.E., A.S.F.B.), Faculty of Pharmacy, Alexandria University, Alexandria, Egypt; Research and Innovation Hub, Alamein International University, Alamein, Egypt (A.F.E.-Y., M.A.E., H.M.H.); and Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar (A.H.E.)
| |
Collapse
|
41
|
Abstract
Recent advances in pharmacotherapies that promote appetite suppression have shown remarkable weight loss. Therapies targeting energy expenditure lag behind, and as such none have yet been identified to be safe and efficacious for sustaining negative energy balance toward weight loss. Multiple energy dissipating pathways have been identified in adipose tissue and muscle. The molecular effectors of some of these pathways have been identified, but much is still left to be learned about their regulation. Understanding the molecular underpinnings of metabolic inefficiency in adipose tissue and muscle is required if these pathways are to be therapeutically targeted in the context of obesity and obesity-accelerated diseases.
Collapse
Affiliation(s)
- Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada
- Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| |
Collapse
|
42
|
Maushart CI, Sun W, Othman A, Ghosh A, Senn JR, Fischer JGW, Madoerin P, Loeliger RC, Benz RM, Takes M, Zech CJ, Chirindel A, Beuschlein F, Reincke M, Wild D, Bieri O, Zamboni N, Wolfrum C, Betz MJ. Effect of high-dose glucocorticoid treatment on human brown adipose tissue activity: a randomised, double-blinded, placebo-controlled cross-over trial in healthy men. EBioMedicine 2023; 96:104771. [PMID: 37659283 PMCID: PMC10483510 DOI: 10.1016/j.ebiom.2023.104771] [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: 02/21/2023] [Revised: 08/07/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Glucocorticoids (GCs) are widely applied anti-inflammatory drugs that are associated with adverse metabolic effects including insulin resistance and weight gain. Previous research indicates that GCs may negatively impact brown adipose tissue (BAT) activity in rodents and humans. METHODS We performed a randomised, double-blinded cross-over trial in 16 healthy men (clinicaltrials.govNCT03269747). Participants received 40 mg of prednisone per day for one week or placebo. After a washout period of four weeks, participants crossed-over to the other treatment arm. Primary endpoint was the increase in resting energy expenditure (EE) in response to a mild-cold stimulus (cold-induced thermogenesis, CIT). Secondary outcomes comprised mean 18F-FDG uptake into supraclavicular BAT (SUVmean) as determined by FDG-PET/CT, volume of the BAT depot as well as fat content determined by MRI. The plasma metabolome and the transcriptome of supraclavicular BAT and of skeletal muscle biopsies after each treatment period were analysed. FINDINGS Sixteen participants were recruited to the trial and completed it successfully per protocol. After prednisone treatment resting EE was higher both during warm and cold conditions. However, CIT was similar, 153 kcal/24 h (95% CI 40-266 kcal/24 h) after placebo and 186 kcal/24 h (95% CI 94-277 kcal/24 h, p = 0.38) after prednisone. SUVmean of BAT after cold exposure was not significantly affected by prednisone (3.36 g/ml, 95% CI 2.69-4.02 g/ml, vs 3.07 g/ml, 95% CI 2.52-3.62 g/ml, p = 0.28). Results of plasma metabolomics and BAT transcriptomics corroborated these findings. RNA sequencing of muscle biopsies revealed higher expression of genes involved in calcium cycling. No serious adverse events were reported and adverse events were evenly distributed between the two treatments. INTERPRETATION Prednisone increased EE in healthy men possibly by altering skeletal muscle calcium cycling. Cold-induced BAT activity was not affected by GC treatment, which indicates that the unfavourable metabolic effects of GCs are independent from thermogenic adipocytes. FUNDING Grants from Swiss National Science Foundation (PZ00P3_167823), Bangerter-Rhyner Foundation and from Nora van der Meeuwen-Häfliger Foundation to MJB. A fellowship-grant from the Swiss National Science Foundation (SNF211053) to WS. Grants from German Research Foundation (project number: 314061271-TRR 205) and Else Kröner-Fresenius (grant support 2012_A103 and 2015_A228) to MR.
Collapse
Affiliation(s)
- Claudia Irene Maushart
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Wenfei Sun
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Alaa Othman
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Adhideb Ghosh
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland; Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Jaël Rut Senn
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Jonas Gabriel William Fischer
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Philipp Madoerin
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Rahel Catherina Loeliger
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| | - Robyn Melanie Benz
- Functional Genomics Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland.
| | - Martin Takes
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Christoph Johannes Zech
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Alin Chirindel
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Felix Beuschlein
- Department of Endocrinology, Diabetology and Clinical Nutrition, University Hospital Zurich (USZ) and University Zurich (UZH), Zurich, Switzerland; Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany.
| | - Martin Reincke
- Department of Medicine IV, University Hospital, LMU Munich, Munich, Germany.
| | - Damian Wild
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Oliver Bieri
- Department of Radiology and Nuclear Medicine, University Hospital Basel, University of Basel, Basel, Switzerland.
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland.
| | - Christian Wolfrum
- Institute of Food, Nutrition, and Health, ETH Zurich, Schwerzenbach, Switzerland.
| | - Matthias Johannes Betz
- Department of Endocrinology, Diabetes and Metabolism, University Hospital Basel and University of Basel, Basel, Switzerland.
| |
Collapse
|
43
|
Ramón A, Esteves A, Villadóniga C, Chalar C, Castro-Sowinski S. A general overview of the multifactorial adaptation to cold: biochemical mechanisms and strategies. Braz J Microbiol 2023; 54:2259-2287. [PMID: 37477802 PMCID: PMC10484896 DOI: 10.1007/s42770-023-01057-4] [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: 03/20/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023] Open
Abstract
Cold environments are more frequent than people think. They include deep oceans, cold lakes, snow, permafrost, sea ice, glaciers, cold soils, cold deserts, caves, areas at elevations greater than 3000 m, and also artificial refrigeration systems. These environments are inhabited by a diversity of eukaryotic and prokaryotic organisms that must adapt to the hard conditions imposed by cold. This adaptation is multifactorial and includes (i) sensing the cold, mainly through the modification of the liquid-crystalline membrane state, leading to the activation of a two-component system that transduce the signal; (ii) adapting the composition of membranes for proper functions mainly due to the production of double bonds in lipids, changes in hopanoid composition, and the inclusion of pigments; (iii) producing cold-adapted proteins, some of which show modifications in the composition of amino acids involved in stabilizing interactions and structural adaptations, e.g., enzymes with high catalytic efficiency; and (iv) producing ice-binding proteins and anti-freeze proteins, extracellular polysaccharides and compatible solutes that protect cells from intracellular and extracellular ice. However, organisms also respond by reprogramming their metabolism and specifically inducing cold-shock and cold-adaptation genes through strategies such as DNA supercoiling, distinctive signatures in promoter regions and/or the action of CSPs on mRNAs, among others. In this review, we describe the main findings about how organisms adapt to cold, with a focus in prokaryotes and linking the information with findings in eukaryotes.
Collapse
Affiliation(s)
- Ana Ramón
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Adriana Esteves
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Carolina Villadóniga
- Laboratorio de Biocatalizadores Y Sus Aplicaciones, Facultad de Ciencias, Instituto de Química Biológica, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Cora Chalar
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay
| | - Susana Castro-Sowinski
- Sección Bioquímica, Instituto de Biología, Facultad de Ciencias, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay.
- Laboratorio de Biocatalizadores Y Sus Aplicaciones, Facultad de Ciencias, Instituto de Química Biológica, Universidad de La República, Igua 4225, 11400, Montevideo, Uruguay.
| |
Collapse
|
44
|
Abstract
In this review, we provide a brief synopsis of the connections between adipose tissue and metabolic health and highlight some recent developments in understanding and exploiting adipocyte biology. Adipose tissue plays critical roles in the regulation of systemic glucose and lipid metabolism and secretes bioactive molecules possessing endocrine, paracrine, and autocrine functions. Dysfunctional adipose tissue has a detrimental impact on metabolic health and is intimately involved in key aspects of metabolic diseases such as insulin resistance, lipid overload, inflammation, and organelle stress. Differences in the distribution of fat depots and adipose characteristics relate to divergent degrees of metabolic dysfunction found in metabolically healthy and unhealthy obese individuals. Thermogenic adipocytes increase energy expenditure via mitochondrial uncoupling or adenosine triphosphate-consuming futile substrate cycles, while functioning as a metabolic sink and participating in crosstalk with other metabolic organs. Manipulation of adipose tissue provides a wealth of opportunities to intervene and combat the progression of associated metabolic diseases. We discuss current treatment modalities for obesity including incretin hormone analogs and touch upon emerging strategies with therapeutic potential including exosome-based therapy, pharmacological activation of brown and beige adipocyte thermogenesis, and administration or inhibition of adipocyte-derived factors.
Collapse
Affiliation(s)
- Sung-Min An
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - Seung-Hee Cho
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| | - John C. Yoon
- Division of Endocrinology, Department of Internal Medicine, University of California Davis School of Medicine, Davis, CA, USA
| |
Collapse
|
45
|
Katsi V, Papakonstantinou I, Tsioufis K. Atherosclerosis, Diabetes Mellitus, and Cancer: Common Epidemiology, Shared Mechanisms, and Future Management. Int J Mol Sci 2023; 24:11786. [PMID: 37511551 PMCID: PMC10381022 DOI: 10.3390/ijms241411786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/03/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
The involvement of cardiovascular disease in cancer onset and development represents a contemporary interest in basic science. It has been recognized, from the most recent research, that metabolic syndrome-related conditions, ranging from atherosclerosis to diabetes, elicit many pathways regulating lipid metabolism and lipid signaling that are also linked to the same framework of multiple potential mechanisms for inducing cancer. Otherwise, dyslipidemia and endothelial cell dysfunction in atherosclerosis may present common or even interdependent changes, similar to oncogenic molecules elevated in many forms of cancer. However, whether endothelial cell dysfunction in atherosclerotic disease provides signals that promote the pre-clinical onset and proliferation of malignant cells is an issue that requires further understanding, even though more questions are presented with every answer. Here, we highlight the molecular mechanisms that point to a causal link between lipid metabolism and glucose homeostasis in metabolic syndrome-related atherosclerotic disease with the development of cancer. The knowledge of these breakthrough mechanisms may pave the way for the application of new therapeutic targets and for implementing interventions in clinical practice.
Collapse
Affiliation(s)
- Vasiliki Katsi
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
| | | | - Konstantinos Tsioufis
- Department of Cardiology, Hippokration Hospital, 11527 Athens, Greece
- School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
46
|
Bienboire-Frosini C, Wang D, Marcet-Rius M, Villanueva-García D, Gazzano A, Domínguez-Oliva A, Olmos-Hernández A, Hernández-Ávalos I, Lezama-García K, Verduzco-Mendoza A, Gómez-Prado J, Mota-Rojas D. The Role of Brown Adipose Tissue and Energy Metabolism in Mammalian Thermoregulation during the Perinatal Period. Animals (Basel) 2023; 13:2173. [PMID: 37443971 DOI: 10.3390/ani13132173] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/21/2023] [Accepted: 06/27/2023] [Indexed: 07/15/2023] Open
Abstract
Hypothermia is one of the most common causes of mortality in neonates, and it could be developed after birth because the uterus temperature is more elevated than the extrauterine temperature. Neonates use diverse mechanisms to thermoregulate, such as shivering and non-shivering thermogenesis. These strategies can be more efficient in some species, but not in others, i.e., altricials, which have the greatest difficulty with achieving thermoneutrality. In addition, there are anatomical and neurological differences in mammals, which may present different distributions and amounts of brown fat. This article aims to discuss the neuromodulation mechanisms of thermoregulation and the importance of brown fat in the thermogenesis of newborn mammals, emphasizing the analysis of the biochemical, physiological, and genetic factors that determine the distribution, amount, and efficiency of this energy resource in newborns of different species. It has been concluded that is vital to understand and minimize hypothermia causes in newborns, which is one of the main causes of mortality in neonates. This would be beneficial for both animals and producers.
Collapse
Affiliation(s)
- Cécile Bienboire-Frosini
- Department of Molecular Biology and Chemical Communication, Research Institute in Semiochemistry and Applied Ethology (IRSEA), 84400 Apt, France
| | - Dehua Wang
- School of Life Sciences, Shandong University, Qingdao 266237, China
| | - Míriam Marcet-Rius
- Animal Behaviour and Welfare Department, Research Institute in Semiochemistry and Applied Ethology (IRSEA), 84400 Apt, France
| | - Dina Villanueva-García
- Division of Neonatology, Hospital Infantil de México Federico Gómez, Mexico City 06720, Mexico
| | - Angelo Gazzano
- Department of Veterinary Sciences, University of Pisa, 56124 Pisa, Italy
| | - Adriana Domínguez-Oliva
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Adriana Olmos-Hernández
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Ismael Hernández-Ávalos
- Clinical Pharmacology and Veterinary Anesthesia, Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Cuautitlán Izcalli 54714, Mexico
| | - Karina Lezama-García
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Antonio Verduzco-Mendoza
- Division of Biotechnology-Bioterio and Experimental Surgery, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra (INR-LGII), Mexico City 14389, Mexico
| | - Jocelyn Gómez-Prado
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| | - Daniel Mota-Rojas
- Neurophysiology, Behavior and Animal Welfare Assessment, DPAA, Universidad Autónoma Metropolitana, Xochimilco Campus, Mexico City 04960, Mexico
| |
Collapse
|
47
|
Nirengi S, Stanford K. Brown adipose tissue and aging: A potential role for exercise. Exp Gerontol 2023; 178:112218. [PMID: 37224933 DOI: 10.1016/j.exger.2023.112218] [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: 09/02/2022] [Revised: 05/05/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Aging is one of the primary risk factors for the development of type 2 diabetes and cardiovascular disease, and regular physical activity can help to delay, prevent, or manage the onset and development of many chronic diseases present in older adults. Brown adipose tissue (BAT) is thermogenic tissue that protects against age-related disease, but BAT activity decreases with age. In this review, we discuss how aging contributes to impaired BAT function by inducing a 'whitening' of the BAT and altering beta 3 adrenergic receptor (β3AR) signaling, uncoupling protein 1 (UCP1) gene expression, and mitochondria respiration, and potential mechanisms for exercise to counteract the effects of aging on BAT.
Collapse
Affiliation(s)
- Shinsuke Nirengi
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Division of Preventive Medicine, Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| | - Kristin Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA; Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
48
|
Klein Hazebroek M, Laterveer R, Kutschke M, Ramšak Marčeta V, Barthem CS, Keipert S. Hyperphagia of female UCP1-deficient mice blunts anti-obesity effects of FGF21. Sci Rep 2023; 13:10288. [PMID: 37355753 PMCID: PMC10290677 DOI: 10.1038/s41598-023-37264-0] [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: 03/20/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023] Open
Abstract
Increasing energy expenditure through uncoupling protein 1 (UCP1) activity in thermogenic adipose tissue is widely investigated to correct diet-induced obesity (DIO). Paradoxically, UCP1-deficient male mice are resistant to DIO at room temperature. Recently, we uncovered a key role for fibroblast growth factor 21 (FGF21), a promising drug target for treatment of metabolic disease, in this phenomenon. As the metabolic action of FGF21 is so far understudied in females, we aim to investigate potential sexual dimorphisms. Here, we confirm that male UCP1 KO mice display resistance to DIO in mild cold, without significant changes in metabolic parameters. Surprisingly, females gained the same amount of body fat as WT controls. Molecular regulation was similar between UCP1 KO males and females, with an upregulation of serum FGF21, coinciding with beiging of inguinal white adipose tissue and induced lipid metabolism. While energy expenditure did not display significant differences, UCP1 KO females significantly increased their food intake. Altogether, our results indicate that hyperphagia is likely counteracting the beneficial effects of FGF21 in female mice. This underlines the importance of sex-specific studies in (pre)clinical research for personalized drug development.
Collapse
Affiliation(s)
- Marlou Klein Hazebroek
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Rutger Laterveer
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Maria Kutschke
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Vida Ramšak Marčeta
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Clarissa S Barthem
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden
| | - Susanne Keipert
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91, Stockholm, Sweden.
| |
Collapse
|
49
|
Tarantini S, Subramanian M, Butcher JT, Yabluchanskiy A, Li X, Miller RA, Balasubramanian P. Revisiting adipose thermogenesis for delaying aging and age-related diseases: Opportunities and challenges. Ageing Res Rev 2023; 87:101912. [PMID: 36924940 PMCID: PMC10164698 DOI: 10.1016/j.arr.2023.101912] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/03/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Adipose tissue undergoes significant changes in structure, composition, and function with age including altered adipokine secretion, decreased adipogenesis, altered immune cell profile and increased inflammation. Considering the role of adipose tissue in whole-body energy homeostasis, age-related dysfunction in adipose metabolism could potentially contribute to an increased risk for metabolic diseases and accelerate the onset of other age-related diseases. Increasing cellular energy expenditure in adipose tissue, also referred to as thermogenesis, has emerged as a promising strategy to improve adipose metabolism and treat obesity-related metabolic disorders. However, translating this strategy to the aged population comes with several challenges such as decreased thermogenic response and the paucity of safe pharmacological agents to activate thermogenesis. This mini-review aims to discuss the current body of knowledge on aging and thermogenesis and highlight the unexplored opportunities (cellular mechanisms and secreted factors) to target thermogenic mechanisms for delaying aging and age-related diseases. Finally, we also discuss the emerging role of thermogenic adipocytes in healthspan and lifespan extension.
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Madhan Subramanian
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Joshua T Butcher
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK, USA
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xinna Li
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Richard A Miller
- Department of Pathology and Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Priya Balasubramanian
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
50
|
Warfel JD, Elks CM, Bayless DS, Vandanmagsar B, Stone AC, Velasquez SE, Olivares-Nazar P, Noland RC, Ghosh S, Zhang J, Mynatt RL. Rats lacking Ucp1 present a novel translational tool for the investigation of thermogenic adaptation during cold challenge. Acta Physiol (Oxf) 2023; 238:e13935. [PMID: 36650072 PMCID: PMC11318575 DOI: 10.1111/apha.13935] [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: 07/05/2022] [Revised: 01/06/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
AIM Valuable studies have tested the role of UCP1 on body temperature maintenance in mice, and we sought to knockout Ucp1 in rats (Ucp1-/- ) to provide insight into thermogenic mechanisms in larger mammals. METHODS We used CRISPR/Cas9 technology to create Ucp1-/- rats. Body weight and adiposity were measured, and rats were subjected to indirect calorimetry. Rats were maintained at room temperature or exposed to 4°C for either 24 h or 14 days. Analyses of brown and white adipose tissue and skeletal muscle were conducted via histology, western blot comparison of oxidative phosphorylation proteins, and qPCR to compare mitochondrial DNA levels and mRNA expression profiles. RNA-seq was performed in skeletal muscle. RESULTS Ucp1-/- rats withstood 4°C for 14 days, but core temperature steadily declined. All rats lost body weight after 14 days at 4°C, but controls increased food intake more robustly than Ucp1-/- rats. Brown adipose tissue showed signs of decreased activity in Ucp1-/- rats, while mitochondrial lipid metabolism markers in white adipose tissue and skeletal muscle were increased. Ucp1-/- rats displayed more visible shivering and energy expenditure than controls at 4°C. Skeletal muscle transcriptomics showed more differences between genotypes at 23°C than at 4°C. CONCLUSION Room temperature presented sufficient cold stress to rats lacking UCP1 to activate compensatory thermogenic mechanisms in skeletal muscle, which were only activated in control rats following exposure to 4°C. These results provide novel insight into thermogenic responses to UCP1 deficiency; and highlight Ucp1-/- rats as an attractive translational model for the study of thermogenesis.
Collapse
Affiliation(s)
- Jaycob D. Warfel
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Carrie M. Elks
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - David S. Bayless
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Bolormaa Vandanmagsar
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Allison C. Stone
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Samuel E. Velasquez
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Paola Olivares-Nazar
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Robert C. Noland
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Sujoy Ghosh
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
- Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - Jingying Zhang
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Randall L. Mynatt
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|