1
|
Shin YC, Latorre-Muro P, Djurabekova A, Zdorevskyi O, Bennett CF, Burger N, Song K, Xu C, Paulo JA, Gygi SP, Sharma V, Liao M, Puigserver P. Structural basis of respiratory complex adaptation to cold temperatures. Cell 2024; 187:6584-6598.e17. [PMID: 39395414 DOI: 10.1016/j.cell.2024.09.029] [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: 02/12/2024] [Revised: 08/07/2024] [Accepted: 09/17/2024] [Indexed: 10/14/2024]
Abstract
In response to cold, mammals activate brown fat for respiratory-dependent thermogenesis reliant on the electron transport chain. Yet, the structural basis of respiratory complex adaptation upon cold exposure remains elusive. Herein, we combined thermoregulatory physiology and cryoelectron microscopy (cryo-EM) to study endogenous respiratory supercomplexes from mice exposed to different temperatures. A cold-induced conformation of CI:III2 (termed type 2) supercomplex was identified with a ∼25° rotation of CIII2 around its inter-dimer axis, shortening inter-complex Q exchange space, and exhibiting catalytic states that favor electron transfer. Large-scale supercomplex simulations in mitochondrial membranes reveal how lipid-protein arrangements stabilize type 2 complexes to enhance catalytic activity. Together, our cryo-EM studies, multiscale simulations, and biochemical analyses unveil the thermoregulatory mechanisms and dynamics of increased respiratory capacity in brown fat at the structural and energetic level.
Collapse
Affiliation(s)
- Young-Cheul Shin
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Pedro Latorre-Muro
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| | - Amina Djurabekova
- Department of Physics, University of Helsinki, Helsinki 00014, Finland
| | | | - Christopher F Bennett
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Nils Burger
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kangkang Song
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Cryo-EM Core Facility, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Chen Xu
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Cryo-EM Core Facility, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Vivek Sharma
- Department of Physics, University of Helsinki, Helsinki 00014, Finland; HiLIFE Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland
| | - Maofu Liao
- Department of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, China; Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, China.
| | - Pere Puigserver
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
2
|
Song SL, Chen XY, Zhao J, Li YY, Xiong YM, Lv L, Chang J, Wang H, Li XH, Qin ZF. Effects of the Fungicide Prothioconazole on Lipid Metabolism in Mice: Whitening Alterations of Brown Adipose Tissue. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2024; 58:18155-18166. [PMID: 39361549 DOI: 10.1021/acs.est.4c05666] [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: 10/05/2024]
Abstract
With considerable concerns about the associations between metabolic disorders and agricultural biocides, there are scattered data suggesting that the triazole fungicide prothioconazole (PTC) at lower doses than the no observed adverse effect level of 5000 μg/kg/d possibly has the potential to disrupt glycolipid metabolism in mammals. Here, we investigated the effects of 50, 500, and 5000 μg/kg/d of PTC on glycolipid metabolism in mice following 8 weeks of administration via drinking water, with specific attention on brown adipose tissue (BAT) and white adipose tissue (WAT) in addition to the liver. We found that along with the increased serum triglyceride level in the 5000 μg/kg/d group, small fatty vacuoles occurred in livers in all treatment groups, indicating lipid accumulation. No change in WAT was observed, but PTC caused BAT whitening, characterized by adipocyte hypertrophy, more unilocular adipocytes with enlarged lipid droplets, reduced UCP1 levels, and down-regulated Doi2 expression, and even the dose of 50 μg/kg/d was effective. Transcriptomic analysis revealed immune inhibition and circadian rhythm disturbance in BAT from the 5000 μg/kg/d group, which are in agreement with BAT whitening and inactivation. On employing the C3H10T1/2 cells in vitro, we found that PTC treatment concentration-dependently promoted lipid accumulation in brown adipocytes, along with altered expression of thermogenesis-related and circadian genes. Taken together, our study shows that low doses of PTC caused BAT whitening, calling for much attention to the new target by pollutants.
Collapse
Affiliation(s)
- Shi-Lin Song
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuan-Yue Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jun Zhao
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Yuan-Yuan Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yi-Ming Xiong
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lin Lv
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Chang
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Huili Wang
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
- Laboratory for Chemical Environmental Risk Assessment, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
| | - Xing Hong Li
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhan-Fen Qin
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
3
|
El-Ansary MRM, El-Ansary AR, Said SM, Abdel-Hakeem MA. Regular cold shower exposure modulates humoral and cell-mediated immunity in healthy individuals. J Therm Biol 2024; 125:103971. [PMID: 39299098 DOI: 10.1016/j.jtherbio.2024.103971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/02/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND Cold hydrotherapy is an ancient practice that has recently gained scientific interest for its potential health benefits. This study explored the effects of regular cold shower exposure on immune cell function. METHODS Sixty healthy Egyptian adults were randomized to take cold or hot showers daily for 90 days. Levels of immunoglobulins, cytokines, and interferon-gamma were measured in blood samples at baseline, 30, 60, and 90 days. RESULTS The cold shower group exhibited significant increases in immunoglobulin levels. Conversely, the hot shower group showed a significant decrease in IgM levels at 60 and 90 days compared to baseline, alongside nonsignificant decrease of IgG and IgA. the cold shower group demonstrated elevated levels of IL-2 and IL-4 at 90 days, indicating enhanced T-cell proliferation and humoral immunity, respectively. In contrast, the hot shower group did not exhibit significant changes in cytokine levels. There were no significant differences in IFN-γ and TNF-α levels between the groups. CONCLUSIONS Regular cold shower exposure appears to enhance humoral and cell-mediated immunity through the upregulation of antibodies, interleukin-2, and interleukin-4. Brief cold stressors may induce physiological adaptations that prime the immune response. This accessible, sustainable lifestyle modification could potentially serve as an alternative therapy to boost immunity. Further research on larger populations is warranted to better understand the physiological effects of cold temperatures on immunity.
Collapse
Affiliation(s)
- Mahmoud R M El-Ansary
- Department of Immunology and Medical Microbiology, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Amira R El-Ansary
- Department of Internal Medicine, Faculty of Medicine, Misr University for Science and Technology, Giza, Egypt
| | - Shereen M Said
- Department of Basic Science, Faculty of Physical Therapy, Misr University for Science and Technology, Giza, Egypt
| | - Mohamed A Abdel-Hakeem
- Department of Pharmaceutical Biotechnology, College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| |
Collapse
|
4
|
Stephens CJ, Kobayashi R, Berry DC, Butcher JT. The Role of Matrix Stiffness And Viscosity on Lipid Phenotype And Fat Lineage Potential. Tissue Eng Part A 2024. [PMID: 39165245 DOI: 10.1089/ten.tea.2024.0149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024] Open
Abstract
Autologous fat transfer is a common procedure that patients undergo to rejuvenate large soft tissue defects. However, these surgeries are complicated by limited tissue sources, donor-site morbidity, and necrosis. While the biofabrication of fat tissue can serve as a clinical option for reconstructive surgery, the influence of matrix mechanics, specifically stiffness and viscosity, on adipogenesis requires further elucidation. Additionally, the effects of these mechanical parameters on metabolic and thermogenic fat potential have yet to be investigated. In this study, gelatin methacryloyl (GelMA) polymers with varying degrees of methacrylation (DoM) were fabricated to create matrices with different stiffnesses and viscosities. Human adipose-derived mesenchymal stem cells were then encapsulated in mechanically tunable GelMA and underwent adipogenesis to investigate the effects of matrix mechanics on lipid phenotype and fat potential. Mechanical testing confirmed that GelMA stiffness was regulated by DoM and weight composition, whereas viscosity was determined by the latter. Further work revealed that while lipid phenotype became more enriched as matrix stiffness and viscosity declined, the potential toward metabolic and thermogenic fat appeared to be more viscous dependent rather than stiffness dependent. In addition, fatty acid binding protein 4 and uncoupling protein 1 gene expression exhibited viscous-dependent behavior despite comparable levels of peroxisome proliferator-activated receptor gamma. However, despite the superior role of viscosity, lipid quantity and mitochondrial abundance demonstrated stiffness-dependent behavior. Overall, this work revealed that matrix viscosity played a more superior role than stiffness in driving adipogenesis and distinguishing between metabolic and thermogenic fat potential. Ultimately, this differentiation in fat production is important for engineering ideal adipose tissue for large soft tissue defects.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Reina Kobayashi
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| | - Daniel C Berry
- College of Human Ecology, Division of Nutrition Science, Cornell University, Ithaca, New York, USA
| | - Jonathan T Butcher
- Cornell University, Nancy E. and Peter C. Meinig School of Biomedical Engineering, Ithaca, New York, USA
| |
Collapse
|
5
|
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
|
6
|
Reynés B, García-Ruiz E, van Schothorst EM, Keijer J, Oliver P, Palou A. TLCD4 as Potential Transcriptomic Biomarker of Cold Exposure. Biomolecules 2024; 14:935. [PMID: 39199323 PMCID: PMC11352221 DOI: 10.3390/biom14080935] [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: 07/01/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 09/01/2024] Open
Abstract
(1) Background: Cold exposure induces metabolic adaptations that can promote health benefits, including increased energy disposal due to lipid mobilization in adipose tissue (AT). This study aims to identify easily measurable biomarkers mirroring the effect of cold exposure on AT. (2) Methods: Transcriptomic analysis was performed in peripheral blood mononuclear cells (PBMCs) and distinct AT depots of two animal models (ferrets and rats) exposed to cold, and in PBMCs of cold-exposed humans. (3) Results: One week of cold exposure (at 4 °C) affected different metabolic pathways and gene expression in the AT of ferrets, an animal model with an AT more similar to humans than that of rodents. However, only one gene, Tlcd4, was affected in the same way (overexpressed) in aortic perivascular and inguinal AT depots and in PBMCs, making it a potential biomarker of interest. Subsequent targeted analysis in rats showed that 1 week at 4 °C also induced Tlcd4 expression in brown AT and PBMCs, while 1 h at 4 °C resulted in reduced Tlcd4 mRNA levels in retroperitoneal white AT. In humans, no clear effects were observed. Nevertheless, decreased PBMC TLCD4 expression was observed after acute cold exposure in women with normal weight, although this effect could be attributed to short-term fasting during the procedure. No effect was evident in women with overweight or in normal-weight men. (4) Conclusions: Our results obtained for different species point toward TLCD4 gene expression as a potential biomarker of cold exposure/fat mobilization that could tentatively be used to address the effectiveness of cold exposure-mimicking therapies.
Collapse
Affiliation(s)
- Bàrbara Reynés
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands, 07122 Palma, Spain; (B.R.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Estefanía García-Ruiz
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands, 07122 Palma, Spain; (B.R.)
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Evert M. van Schothorst
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands; (E.M.v.S.)
| | - Jaap Keijer
- Human and Animal Physiology, Wageningen University, 6708 WD Wageningen, The Netherlands; (E.M.v.S.)
| | - Paula Oliver
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands, 07122 Palma, Spain; (B.R.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Andreu Palou
- Nutrigenomics, Biomarkers and Risk Evaluation (NuBE) Group, University of the Balearic Islands, 07122 Palma, Spain; (B.R.)
- Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
- CIBER of Physiopathology of Obesity and Nutrition (CIBEROBN), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
7
|
Ishida Y, Matsushita M, Yoneshiro T, Saito M, Fuse S, Hamaoka T, Kuroiwa M, Tanaka R, Kurosawa Y, Nishimura T, Motoi M, Maeda T, Nakayama K. Genetic evidence for involvement of β2-adrenergic receptor in brown adipose tissue thermogenesis in humans. Int J Obes (Lond) 2024; 48:1110-1117. [PMID: 38632325 PMCID: PMC11281906 DOI: 10.1038/s41366-024-01522-6] [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: 12/02/2023] [Revised: 04/05/2024] [Accepted: 04/08/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Sympathetic activation of brown adipose tissue (BAT) thermogenesis can ameliorate obesity and related metabolic abnormalities. However, crucial subtypes of the β-adrenergic receptor (AR), as well as effects of its genetic variants on functions of BAT, remains unclear in humans. We conducted association analyses of genes encoding β-ARs and BAT activity in human adults. METHODS Single nucleotide polymorphisms (SNPs) in β1-, β2-, and β3-AR genes (ADRB1, ADRB2, and ADRB3) were tested for the association with BAT activity under mild cold exposure (19 °C, 2 h) in 399 healthy Japanese adults. BAT activity was measured using fluorodeoxyglucose-positron emission tomography and computed tomography (FDG-PET/CT). To validate the results, we assessed the effects of SNPs in the two independent populations comprising 277 healthy East Asian adults using near-infrared time-resolved spectroscopy (NIRTRS) or infrared thermography (IRT). Effects of SNPs on physiological responses to intensive cold exposure were tested in 42 healthy Japanese adult males using an artificial climate chamber. RESULTS We found a significant association between a functional SNP (rs1042718) in ADRB2 and BAT activity assessed with FDG-PET/CT (p < 0.001). This SNP also showed an association with cold-induced thermogenesis in the population subset. Furthermore, the association was replicated in the two other independent populations; BAT activity was evaluated by NIRTRS or IRT (p < 0.05). This SNP did not show associations with oxygen consumption and cold-induced thermogenesis under intensive cold exposure, suggesting the irrelevance of shivering thermogenesis. The SNPs of ADRB1 and ADRB3 were not associated with these BAT-related traits. CONCLUSIONS The present study supports the importance of β2-AR in the sympathetic regulation of BAT thermogenesis in humans. The present collection of DNA samples is the largest to which information on the donor's BAT activity has been assigned and can serve as a reference for further in-depth understanding of human BAT function.
Collapse
MESH Headings
- Humans
- Thermogenesis/physiology
- Thermogenesis/genetics
- Adipose Tissue, Brown/metabolism
- Male
- Adult
- Polymorphism, Single Nucleotide
- Receptors, Adrenergic, beta-2/genetics
- Receptors, Adrenergic, beta-2/metabolism
- Female
- Middle Aged
- Japan
- Positron Emission Tomography Computed Tomography
- Receptors, Adrenergic, beta-3/genetics
- Receptors, Adrenergic, beta-3/metabolism
- Asian People/genetics
Collapse
Affiliation(s)
- Yuka Ishida
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065-0013, Japan
| | - Takeshi Yoneshiro
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro-ku, Tokyo, 153-8904, Japan
- Department of Molecular Metabolism and Physiology, Graduate School of Medicine, Tohoku University, Aoba-ku, Sendai, 980-8575, Japan
| | - Masayuki Saito
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065-0013, Japan
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido, 060-0818, Japan
| | - Sayuri Fuse
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takafumi Hamaoka
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Miyuki Kuroiwa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Riki Tanaka
- Faculty of Sports and Health Science, Fukuoka University, Fukuoka, Fukuoka, 814-0180, Japan
| | - Yuko Kurosawa
- Department of Sports Medicine for Health Promotion, Tokyo Medical University, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Takayuki Nishimura
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
- Physiological Anthropology Research Center, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Midori Motoi
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Takafumi Maeda
- Department of Human Life Design and Science, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
- Physiological Anthropology Research Center, Faculty of Design, Kyushu University, Fukuoka, Fukuoka, 815-8540, Japan
| | - Kazuhiro Nakayama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, 277-8562, Japan.
| |
Collapse
|
8
|
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
|
9
|
Langer HT, Rohm M, Goncalves MD, Sylow L. AMPK as a mediator of tissue preservation: time for a shift in dogma? Nat Rev Endocrinol 2024:10.1038/s41574-024-00992-y. [PMID: 38760482 DOI: 10.1038/s41574-024-00992-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 05/19/2024]
Abstract
Ground-breaking discoveries have established 5'-AMP-activated protein kinase (AMPK) as a central sensor of metabolic stress in cells and tissues. AMPK is activated through cellular starvation, exercise and drugs by either directly or indirectly affecting the intracellular AMP (or ADP) to ATP ratio. In turn, AMPK regulates multiple processes of cell metabolism, such as the maintenance of cellular ATP levels, via the regulation of fatty acid oxidation, glucose uptake, glycolysis, autophagy, mitochondrial biogenesis and degradation, and insulin sensitivity. Moreover, AMPK inhibits anabolic processes, such as lipogenesis and protein synthesis. These findings support the notion that AMPK is a crucial regulator of cell catabolism. However, studies have revealed that AMPK's role in cell homeostasis might not be as unidirectional as originally thought. This Review explores emerging evidence for AMPK as a promoter of cell survival and an enhancer of anabolic capacity in skeletal muscle and adipose tissue during catabolic crises. We discuss AMPK-activating interventions for tissue preservation during tissue wasting in cancer-associated cachexia and explore the clinical potential of AMPK activation in wasting conditions. Overall, we provide arguments that call for a shift in the current dogma of AMPK as a mere regulator of cell catabolism, concluding that AMPK has an unexpected role in tissue preservation.
Collapse
Affiliation(s)
- Henning Tim Langer
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA.
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riβ, Germany.
| | - Maria Rohm
- Institute for Diabetes and Cancer, Helmholtz Center Munich, Neuherberg, Germany
- Joint Heidelberg-IDC Translational Diabetes Program, Inner Medicine 1, Heidelberg University Hospital, Heidelberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Marcus DaSilva Goncalves
- Division of Endocrinology, Weill Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Lykke Sylow
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Henckel MM, Chun JH, Knaub LA, Pott GB, James GE, Hunter KS, Shandas R, Walker LA, Reusch JEB, Keller AC. Perivascular adipose tissue remodeling impairs vasoreactivity in thermoneutral-housed rats. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.09.593330. [PMID: 38798439 PMCID: PMC11118269 DOI: 10.1101/2024.05.09.593330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Objective Vascular pathology, characterized by impaired vasoreactivity and mitochondrial respiration, differs between the sexes. Housing rats under thermoneutral (TN) conditions causes vascular dysfunction and perturbed metabolism. We hypothesized that perivascular adipose tissue (PVAT), a vasoregulatory adipose depot with brown adipose tissue (BAT) phenotype, remodels to a white adipose (WAT) phenotype in rats housed at TN, driving diminished vasoreactivity in a sex-dependent manner. Methods Male and female Wistar rats were housed at either room temperature (RT) or TN. Endpoints included changes in PVAT morphology, vasoreactivity in vessels with intact PVAT or transferred to PVAT of the oppositely-housed animal, vessel stiffness, vessel mitochondrial respiration and cellular signaling. Results Remodeling of PVAT was observed in rats housed at TN; animals in this environment showed PVAT whitening and displayed diminished aortae vasodilation (p<0.05), different between the sexes. Juxtaposing PVAT from RT rats onto aortae from TN rats in females corrected vasodilation (p<0.05); this did not occur in males. In aortae of all animals housed at TN, mitochondrial respiration was significantly diminished in lipid substrate experiments (p<0.05), and there was significantly less expression of peNOS (p<0.001). Conclusions These data are consistent with TN-induced remodeling of PVAT, notably associated with sex-specific blunting of vasoreactivity, diminished mitochondrial respiration, and altered cellular signaling.
Collapse
Affiliation(s)
- Melissa M Henckel
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Ji Hye Chun
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Leslie A Knaub
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Gregory B Pott
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | | | - Kendall S Hunter
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Robin Shandas
- Department of Bioengineering, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Lori A Walker
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Jane E-B Reusch
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| | - Amy C Keller
- Division of Endocrinology, Metabolism & Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
- Rocky Mountain Regional VA Medical Center, Aurora, CO 80045
| |
Collapse
|
11
|
Dumont L, Caron A, Richard G, Croteau E, Fortin M, Frisch F, Phoenix S, Dubreuil S, Guérin B, Turcotte ÉE, Carpentier AC, Blondin DP. The effects of the β 1-adrenergic receptor antagonist bisoprolol administration on mirabegron-stimulated human brown adipose tissue thermogenesis. Acta Physiol (Oxf) 2024; 240:e14127. [PMID: 38502056 DOI: 10.1111/apha.14127] [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: 09/02/2023] [Revised: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
AIM Pharmacological stimulation of human brown adipose tissue (BAT) has been hindered by ineffective activation or undesirable off-target effects. Oral administration of the maximal allowable dose of mirabegron (200 mg), a β3-adrenergic receptor (β3-AR) agonist, has been effective in stimulating BAT thermogenesis and whole-body energy expenditure. However, this has been accompanied by undesirable cardiovascular effects. Therefore, we hypothesized that combining mirabegron with a β1-AR antagonist could suppress these unwanted effects and increase the stimulation of the β3-AR and β2-AR in BAT. METHODS We performed a randomized crossover trial (NCT04823442) in 8 lean men. Mirabegron (200 mg) was administered orally with or without the β1-AR antagonist bisoprolol (10 mg). Dynamic [11C]-acetate and 2-deoxy-2-[18F]fluoro-d-glucose PET/CT scans were performed sequentially after oral administration of mirabegron ± bisoprolol. RESULTS Compared to room temperature, mirabegron alone increased BAT oxidative metabolism (0.84 ± 0.46 vs. 1.79 ± 0.91 min-1, p = 0.0433), but not when combined with bisoprolol. The metabolic rate of glucose in BAT, measured using [18F]FDG PET, was significantly higher with mirabegron than mirabegron with bisoprolol (24 ± 10 vs. 16 ± 8 nmol/g/min, p = 0.0284). Bisoprolol inhibited the mirabegron-induced increase in systolic blood pressure and heart rate. CONCLUSION The administration of bisoprolol decreases the adverse cardiovascular effects of mirabegron. However, the provided dose also blunted the mirabegron-stimulated increase in BAT lipolysis, thermogenesis, and glucose uptake. The attenuation in BAT blood flow induced by the large dose of bisoprolol may have limited BAT thermogenesis.
Collapse
Affiliation(s)
- Lauralyne Dumont
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Department of Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, Québec, Canada
- Quebec Heart and Lung Institute, Quebec City, Québec, Canada
- Montreal Diabetes Research Center, Montreal, Québec, Canada
| | - Gabriel Richard
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Etienne Croteau
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Mélanie Fortin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Frédérique Frisch
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Serge Phoenix
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Stéphanie Dubreuil
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Brigitte Guérin
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Éric E Turcotte
- Centre d'Imagerie Moléculaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - André C Carpentier
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Denis P Blondin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
- Division of Neurology, Department of Medicine, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
12
|
Negroiu CE, Tudorașcu I, Bezna CM, Godeanu S, Diaconu M, Danoiu R, Danoiu S. Beyond the Cold: Activating Brown Adipose Tissue as an Approach to Combat Obesity. J Clin Med 2024; 13:1973. [PMID: 38610736 PMCID: PMC11012454 DOI: 10.3390/jcm13071973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 03/18/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
With a dramatic increase in the number of obese and overweight people, there is a great need for new anti-obesity therapies. With the discovery of the functionality of brown adipose tissue in adults and the observation of beige fat cells among white fat cells, scientists are looking for substances and methods to increase the activity of these cells. We aimed to describe how scientists have concluded that brown adipose tissue is also present and active in adults, to describe where in the human body these deposits of brown adipose tissue are, to summarize the origin of both brown fat cells and beige fat cells, and, last but not least, to list some of the substances and methods classified as BAT promotion agents with their benefits and side effects. We summarized these findings based on the original literature and reviews in the field, emphasizing the discovery, function, and origins of brown adipose tissue, BAT promotion agents, and batokines. Only studies written in English and with a satisfying rating were identified from electronic searches of PubMed.
Collapse
Affiliation(s)
- Cristina Elena Negroiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (C.M.B.); (S.D.)
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | - Iulia Tudorașcu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (C.M.B.); (S.D.)
| | - Cristina Maria Bezna
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (C.M.B.); (S.D.)
| | - Sanziana Godeanu
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
- Department of Physiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Marina Diaconu
- Department of Radiology, County Clinical Emergency Hospital of Craiova, 200642 Craiova, Romania;
| | - Raluca Danoiu
- Department of Social Sciences and Humanities, University of Craiova, 200585 Craiova, Romania;
| | - Suzana Danoiu
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania; (C.M.B.); (S.D.)
| |
Collapse
|
13
|
Niclou A, Vesi L, Arorae M, Naseri NC, Savusa KF, Naseri T, Young J, Rivara AC, Ocobock C. Indication of mixed glucose and fatty acid use by inferred brown adipose tissue activity in Samoans. Am J Hum Biol 2024; 36:e23998. [PMID: 37823535 PMCID: PMC10939975 DOI: 10.1002/ajhb.23998] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 09/11/2023] [Accepted: 09/21/2023] [Indexed: 10/13/2023] Open
Abstract
OBJECTIVES Despite the growing rates of global obesity and the known positive associations between brown adipose tissue (BAT) and cardiovascular health, little is known about the metabolic effects of BAT activity in Samoans, a population at high risk of obesity and type II diabetes. Here we assessed the potential effects of inferred BAT activity on metabolic health markers in Samoan adults exposed to mild cold. METHODS Using point-of-care finger prick technology we measured fasting glucose, total cholesterol, high-density lipoprotein (HDL), and low-density lipoprotein (LDL) levels before and after 30 min of cold exposure among 61 individuals (38 females, 23 males, ages 31-54) from 'Upolu Island, Samoa. Respiratory quotient was measured by indirect calorimetry to determine substrate metabolism at room temperature and cold exposure. RESULTS Fasting glucose levels decreased significantly (p < .001) after cold exposure while neither total cholesterol (p = .88), HDL (p = .312), nor LDL (p = .089) changed. Respiratory quotient decreased significantly (p = .009) between exposures, suggesting an increased preference for lipid metabolism as a response to cold. CONCLUSIONS The observed effects of inferred BAT activity on biomarkers suggest BAT activity utilizes both glucose and lipid-derived fatty acids as fuel for thermogenesis. Our work provides evidence for the beneficial metabolic effects of BAT and emphasizes the need for the population-specific development of metabolic treatments involving BAT to ensure the successful and equitable minimization of extreme consequences of obesity and metabolic health.
Collapse
Affiliation(s)
- Alexandra Niclou
- Pennington Biomedical Research Center, Baton Rouge, LA
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
| | - Lupesina Vesi
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | - Maria Arorae
- Obesity, Lifestyle and Genetic Adaptations (OLaGA) Study Group, Apia, Samoa
| | | | | | | | - Jessica Young
- Center for Social Science Research, University of Notre Dame, Notre Dame, IN
| | - Anna C. Rivara
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, CT
| | - Cara Ocobock
- Department of Anthropology, University of Notre Dame, Notre Dame, IN
- Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN
| |
Collapse
|
14
|
Mota CMD, Madden CJ. Neural circuits of long-term thermoregulatory adaptations to cold temperatures and metabolic demands. Nat Rev Neurosci 2024; 25:143-158. [PMID: 38316956 DOI: 10.1038/s41583-023-00785-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2023] [Indexed: 02/07/2024]
Abstract
The mammalian brain controls heat generation and heat loss mechanisms that regulate body temperature and energy metabolism. Thermoeffectors include brown adipose tissue, cutaneous blood flow and skeletal muscle, and metabolic energy sources include white adipose tissue. Neural and metabolic pathways modulating the activity and functional plasticity of these mechanisms contribute not only to the optimization of function during acute challenges, such as ambient temperature changes, infection and stress, but also to longitudinal adaptations to environmental and internal changes. Exposure of humans to repeated and seasonal cold ambient conditions leads to adaptations in thermoeffectors such as habituation of cutaneous vasoconstriction and shivering. In animals that undergo hibernation and torpor, neurally regulated metabolic and thermoregulatory adaptations enable survival during periods of significant reduction in metabolic rate. In addition, changes in diet can activate accessory neural pathways that alter thermoeffector activity. This knowledge may be harnessed for therapeutic purposes, including treatments for obesity and improved means of therapeutic hypothermia.
Collapse
Affiliation(s)
- Clarissa M D Mota
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Christopher J Madden
- Department of Neurological Surgery, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
15
|
Wang T, Zhao C, Zhang J, Li S, Zhang Y, Gong Y, Zhou Y, Yan L, Zhang S, Zhang Z, Hu H, Liu A, Bai X, Zou Z. Whitening of brown adipose tissue inhibits osteogenic differentiation via secretion of S100A8/A9. iScience 2024; 27:108857. [PMID: 38303710 PMCID: PMC10830855 DOI: 10.1016/j.isci.2024.108857] [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: 07/18/2023] [Revised: 11/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
The mechanism by which brown adipose tissue (BAT) regulates bone metabolism is unclear. Here, we reveal that BAT secretes S100A8/A9, a previously unidentified BAT adipokine (batokine), to impair bone formation. Brown adipocytes-specific knockout of Rheb (RhebBAD KO), the upstream activator of mTOR, causes BAT malfunction to inhibit osteogenesis. Rheb depletion induces NF-κB dependent S100A8/A9 secretion from brown adipocytes, but not from macrophages. In wild-type mice, age-related Rheb downregulation in BAT is associated with enhanced S100A8/A9 secretion. Either batokines from RhebBAD KO mice, or recombinant S100A8/A9, inhibits osteoblast differentiation of mesenchymal stem cells in vitro by targeting toll-like receptor 4 on their surfaces. Conversely, S100A8/A9 neutralization not only rescues the osteogenesis repressed in the RhebBAD KO mice, but also alleviates age-related osteoporosis in wild-type mice. Collectively, our data revealed an unexpected BAT-bone crosstalk driven by Rheb-S100A8/A9, uncovering S100A8/A9 as a promising target for the treatment, and potentially, prevention of osteoporosis.
Collapse
Affiliation(s)
- Ting Wang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Chaoran Zhao
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jiahuan Zhang
- Laboratory Medicine, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shengfa Li
- Clinical Research Center, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Youming Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Gong
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yingyue Zhou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lei Yan
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Sheng Zhang
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopadics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hongling Hu
- Department of Trauma and Joint Surgery, Shunde Hospital, Southern Medical University, Foshan, China
| | - Anling Liu
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhipeng Zou
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
16
|
Ronayne CT, Latorre-Muro P. Navigating the landscape of mitochondrial-ER communication in health and disease. Front Mol Biosci 2024; 11:1356500. [PMID: 38323074 PMCID: PMC10844478 DOI: 10.3389/fmolb.2024.1356500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 01/10/2024] [Indexed: 02/08/2024] Open
Abstract
Intracellular organelle communication enables the maintenance of tissue homeostasis and health through synchronized adaptive processes triggered by environmental cues. Mitochondrial-Endoplasmic Reticulum (ER) communication sustains cellular fitness by adjusting protein synthesis and degradation, and metabolite and protein trafficking through organelle membranes. Mitochondrial-ER communication is bidirectional and requires that the ER-components of the Integrated Stress Response signal to mitochondria upon activation and, likewise, mitochondria signal to the ER under conditions of metabolite and protein overload to maintain proper functionality and ensure cellular survival. Declines in the mitochondrial-ER communication occur upon ageing and correlate with the onset of a myriad of heterogeneous age-related diseases such as obesity, type 2 diabetes, cancer, or neurodegenerative pathologies. Thus, the exploration of the molecular mechanisms of mitochondrial-ER signaling and regulation will provide insights into the most fundamental cellular adaptive processes with important therapeutical opportunities. In this review, we will discuss the pathways and mechanisms of mitochondrial-ER communication at the mitochondrial-ER interface and their implications in health and disease.
Collapse
Affiliation(s)
- Conor T. Ronayne
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, United States
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Shin YC, Latorre-Muro P, Djurabekova A, Zdorevskyi O, Bennett CF, Burger N, Song K, Xu C, Sharma V, Liao M, Puigserver P. Structural basis of respiratory complexes adaptation to cold temperatures. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.16.575914. [PMID: 38293190 PMCID: PMC10827213 DOI: 10.1101/2024.01.16.575914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
In response to cold, mammals activate brown fat for respiratory-dependent thermogenesis reliant on the electron transport chain (1, 2). Yet, the structural basis of respiratory complex adaptation to cold remains elusive. Herein we combined thermoregulatory physiology and cryo-EM to study endogenous respiratory supercomplexes exposed to different temperatures. A cold-induced conformation of CI:III 2 (termed type 2) was identified with a ∼25° rotation of CIII 2 around its inter-dimer axis, shortening inter-complex Q exchange space, and exhibiting different catalytic states which favor electron transfer. Large-scale supercomplex simulations in lipid membrane reveal how unique lipid-protein arrangements stabilize type 2 complexes to enhance catalytic activity. Together, our cryo-EM studies, multiscale simulations and biochemical analyses unveil the mechanisms and dynamics of respiratory adaptation at the structural and energetic level.
Collapse
|
18
|
Carpentier AC, Blondin DP. Human brown adipose tissue is not enough to combat cardiometabolic diseases. J Clin Invest 2023; 133:e175288. [PMID: 38038130 PMCID: PMC10688973 DOI: 10.1172/jci175288] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023] Open
Affiliation(s)
| | - Denis P. Blondin
- Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| |
Collapse
|
19
|
Solianik R, Židonienė K, Brazaitis M. Short-duration cold exposure decreases fasting-induced glucose intolerance but has no effect on resting energy expenditure. Cryobiology 2023; 113:104564. [PMID: 37541564 DOI: 10.1016/j.cryobiol.2023.104564] [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: 03/26/2023] [Revised: 06/19/2023] [Accepted: 08/01/2023] [Indexed: 08/06/2023]
Abstract
The aim of the present study was to investigate whether brief cold exposure can reverse fasting-induced glucose intolerance and insulin resistance, and improve resting energy expenditure (REE). Twelve young non-obese women were randomly assigned to undergo the following conditions: 2 days of fasting with two 10-min whole-body cold-water immersions on separate days (FAST-COLD), 2 days of fasting without cold-water immersions (FAST), 2 days of usual diet with two 10-min whole-body cold-water immersions on separate days (COLD), or 2 days of usual diet without cold-water immersions (CON) in a randomised crossover fashion. Changes in REE and substrate utilisation, and glucose tolerance and insulin sensitivity from the oral glucose tolerance test were examined. The results showed that FAST-COLD and FAST trials increased (P < 0.05) REE and decreased (P < 0.05) respiratory quotient, but these variables did not differ significantly between the FAST-COLD and FAST trials. The glucose and insulin area under the curves (AUCs) were higher (P < 0.05) in the FAST-COLD and FAST trials than in the CON and COLD trials, and these AUCs were lower (P < 0.05) in the FAST-COLD than in the FAST trial. Matsuda index was lower in the FAST trial than in the CON trial (P < 0.05), and tended to be greater after the FAST-COLD trial than after the FAST trial (P = 0.060). In conclusion, cold exposure had no effect on REE but decreased fasting-induced glucose intolerance which was accompanied by a maintained insulin sensitivity.
Collapse
Affiliation(s)
- Rima Solianik
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania.
| | - Katerina Židonienė
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - Marius Brazaitis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
20
|
Zhang P, Liao D, Yu H, Qiu H. Innovative Metabolic Rate Sensing Approach for Probing Human Thermal Comfort. ACS Biomater Sci Eng 2023; 9:6504-6514. [PMID: 37796119 DOI: 10.1021/acsbiomaterials.3c01029] [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] [Indexed: 10/06/2023]
Abstract
The human metabolic rate has attracted increasing interest as it is the most critical parameter in thermal comfort evaluation, a challenging field, while it is always determined imprecisely. The main issue hampering metabolic rate portable measurement is a lack of reliable methods. Current measuring solutions are unsatisfactory because nonportable bulky size systems and disturbance masks are required. This paper proposes a novel metabolic rate measurement model, which we believe is the first of its kind, to accurately identify and predict human metabolism values via wearable technology. Based on a newly developed theory, the designed wearable metabolic rate sensor was fabricated to measure key parameters: heart rate, heat loss, and skin resistance. Together with the body muscle rate, the new final linear metabolic rate model showed easy prediction capability. Eight volunteers were invited for the experiment under three conditions under four activity intensity states. First, the results significantly verify that a linear relationship exists between the metabolic rate tested by the Quark CPET instrument and our proposed model, with a high coefficient of determination (R2 ≈ 0.90). The correlation model is worth mentioning because it coincides with our hypothesis, with at least 95% overall accuracy and less than 2% uncertainty under each condition. Second, the most remarkable finding is that our model is exceedingly suitable (R2 ≈ 0.90) for the same person, regardless of the experimental temperature. Finally, validation is conducted in a wider metabolic range, further strengthening confidence in our metabolic rate estimation approach. In summary, based on an innovative methodology, our novel metabolic rate sensor is wearable, comfortable, real-time achievable, and miniaturized compared with the existing equipment. This paper sheds new light on human metabolic rate measurement and prediction. Furthermore, our approach and designed sensor can be applied to evaluate indoor thermal comfort precisely, thus leading to reduced energy consumption.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Dong Liao
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Hongyu Yu
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Huihe Qiu
- Department of Mechanical and Aerospace Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
- Thrust of Sustainable Energy and Environment, The Hong Kong University of Science and Technology (Guangzhou), Guangzhou 511400, China
| |
Collapse
|
21
|
Bornstein MR, Neinast MD, Zeng X, Chu Q, Axsom J, Thorsheim C, Li K, Blair MC, Rabinowitz JD, Arany Z. Comprehensive quantification of metabolic flux during acute cold stress in mice. Cell Metab 2023; 35:2077-2092.e6. [PMID: 37802078 PMCID: PMC10840821 DOI: 10.1016/j.cmet.2023.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/14/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023]
Abstract
Cold-induced thermogenesis (CIT) is widely studied as a potential avenue to treat obesity, but a thorough understanding of the metabolic changes driving CIT is lacking. Here, we present a comprehensive and quantitative analysis of the metabolic response to acute cold exposure, leveraging metabolomic profiling and minimally perturbative isotope tracing studies in unanesthetized mice. During cold exposure, brown adipose tissue (BAT) primarily fueled the tricarboxylic acid (TCA) cycle with fat in fasted mice and glucose in fed mice, underscoring BAT's metabolic flexibility. BAT minimally used branched-chain amino acids or ketones, which were instead avidly consumed by muscle during cold exposure. Surprisingly, isotopic labeling analyses revealed that BAT uses glucose largely for TCA anaplerosis via pyruvate carboxylation. Finally, we find that cold-induced hepatic gluconeogenesis is critical for CIT during fasting, demonstrating a key functional role for glucose metabolism. Together, these findings provide a detailed map of the metabolic rewiring driving acute CIT.
Collapse
Affiliation(s)
- Marc R Bornstein
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael D Neinast
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Xianfeng Zeng
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Qingwei Chu
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jessie Axsom
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Chelsea Thorsheim
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kristina Li
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Megan C Blair
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua D Rabinowitz
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Zoltan Arany
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Cero C, Shu W, Reese AL, Douglas D, Maddox M, Singh AP, Ali SL, Zhu AR, Katz JM, Pierce AE, Long KT, Nilubol N, Cypess RH, Jacobs JL, Tian F, Cypess AM. Standardized In Vitro Models of Human Adipose Tissue Reveal Metabolic Flexibility in Brown Adipocyte Thermogenesis. Endocrinology 2023; 164:bqad161. [PMID: 37944134 PMCID: PMC11032247 DOI: 10.1210/endocr/bqad161] [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: 06/05/2023] [Revised: 10/10/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
Functional human brown and white adipose tissue (BAT and WAT) are vital for thermoregulation and nutritional homeostasis, while obesity and other stressors lead, respectively, to cold intolerance and metabolic disease. Understanding BAT and WAT physiology and dysfunction necessitates clinical trials complemented by mechanistic experiments at the cellular level. These require standardized in vitro models, currently lacking, that establish references for gene expression and function. We generated and characterized a pair of immortalized, clonal human brown (hBA) and white (hWA) preadipocytes derived from the perirenal and subcutaneous depots, respectively, of a 40-year-old male individual. Cells were immortalized with hTERT and confirmed to be of a mesenchymal, nonhematopoietic lineage based on fluorescence-activated cell sorting and DNA barcoding. Functional assessments showed that the hWA and hBA phenocopied primary adipocytes in terms of adrenergic signaling, lipolysis, and thermogenesis. Compared to hWA, hBA were metabolically distinct, with higher rates of glucose uptake and lactate metabolism, and greater basal, maximal, and nonmitochondrial respiration, providing a mechanistic explanation for the association between obesity and BAT dysfunction. The hBA also responded to the stress of maximal respiration by using both endogenous and exogenous fatty acids. In contrast to certain mouse models, hBA adrenergic thermogenesis was mediated by several mechanisms, not principally via uncoupling protein 1 (UCP1). Transcriptomics via RNA-seq were consistent with the functional studies and established a molecular signature for each cell type before and after differentiation. These standardized cells are anticipated to become a common resource for future physiological, pharmacological, and genetic studies of human adipocytes.
Collapse
Affiliation(s)
- Cheryl Cero
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Weiguo Shu
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Amy L Reese
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Diana Douglas
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - Michael Maddox
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
- Current Affiliation: Vita Therapeutics, 801 W. Baltimore Street, Suite 301, Baltimore, MD 21201, USA
| | - Ajeet P Singh
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Sahara L Ali
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alexander R Zhu
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jacqueline M Katz
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anne E Pierce
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kelly T Long
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naris Nilubol
- Surgical Oncology Program, Center for Cancer Research, NCI, NIH, 10 Center Drive, Room 4-5952, Bethesda, MD 20892, USA
| | - Raymond H Cypess
- American Type Culture Collection, 10801 University Blvd, Manassas, VA 20110, USA
| | - Jonathan L Jacobs
- American Type Culture Collection, Sequencing and Bioinformatics Center, 10801 University Blvd, Manassas, VA 20110, USA
| | - Fang Tian
- American Type Culture Collection, Cell Biology R&D, 217 Perry Parkway, Gaithersburg, MD 20877, USA
| | - 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
| |
Collapse
|
23
|
Genchi VA, Palma G, Sorice GP, D'Oria R, Caccioppoli C, Marrano N, Biondi G, Caruso I, Cignarelli A, Natalicchio A, Laviola L, Giorgino F, Perrini S. Pharmacological modulation of adaptive thermogenesis: new clues for obesity management? J Endocrinol Invest 2023; 46:2213-2236. [PMID: 37378828 PMCID: PMC10558388 DOI: 10.1007/s40618-023-02125-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 05/29/2023] [Indexed: 06/29/2023]
Abstract
BACKGROUND Adaptive thermogenesis represents the main mechanism through which the body generates heat in response to external stimuli, a phenomenon that includes shivering and non-shivering thermogenesis. The non-shivering thermogenesis is mainly exploited by adipose tissue characterized by a brown aspect, which specializes in energy dissipation. A decreased amount of brown adipose tissue has been observed in ageing and chronic illnesses such as obesity, a worldwide health problem characterized by dysfunctional adipose tissue expansion and associated cardiometabolic complications. In the last decades, the discovery of a trans-differentiation mechanism ("browning") within white adipose tissue depots, leading to the generation of brown-like cells, allowed to explore new natural and synthetic compounds able to favour this process and thus enhance thermogenesis with the aim of counteracting obesity. Based on recent findings, brown adipose tissue-activating agents could represent another option in addition to appetite inhibitors and inhibitors of nutrient absorption for obesity treatment. PURPOSE This review investigates the main molecules involved in the physiological (e.g. incretin hormones) and pharmacological (e.g. β3-adrenergic receptors agonists, thyroid receptor agonists, farnesoid X receptor agonists, glucagon-like peptide-1, and glucagon receptor agonists) modulation of adaptive thermogenesis and the signalling mechanisms involved.
Collapse
Affiliation(s)
- V A Genchi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Palma
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G P Sorice
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - R D'Oria
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - C Caccioppoli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - N Marrano
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - G Biondi
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - I Caruso
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Cignarelli
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - A Natalicchio
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - L Laviola
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| | - F Giorgino
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy.
| | - S Perrini
- Section of Internal Medicine, Endocrinology, Andrology and Metabolic Diseases, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare, 11, 70124, Bari, Italy
| |
Collapse
|
24
|
He M, Li Z, Tung VSK, Pan M, Han X, Evgrafov O, Jiang XC. Inhibiting Phosphatidylcholine Remodeling in Adipose Tissue Increases Insulin Sensitivity. Diabetes 2023; 72:1547-1559. [PMID: 37625119 PMCID: PMC10588299 DOI: 10.2337/db23-0317] [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: 04/25/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023]
Abstract
Cell membrane phosphatidylcholine (PC) composition is regulated by lysophosphatidylcholine acyltransferase (LPCAT); changes in membrane PC saturation are implicated in metabolic disorders. Here, we identified LPCAT3 as the major isoform of LPCAT in adipose tissue and created adipocyte-specific Lpcat3-knockout mice to study adipose tissue lipid metabolism. Transcriptome sequencing and plasma adipokine profiling were used to investigate how LPCAT3 regulates adipose tissue insulin signaling. LPCAT3 deficiency reduced polyunsaturated PCs in adipocyte plasma membranes, increasing insulin sensitivity. LPCAT3 deficiency influenced membrane lipid rafts, which activated insulin receptors and AKT in adipose tissue, and attenuated diet-induced insulin resistance. Conversely, higher LPCAT3 activity in adipose tissue from ob/ob, db/db, and high-fat diet-fed mice reduced insulin signaling. Adding polyunsaturated PCs to mature human or mouse adipocytes in vitro worsened insulin signaling. We suggest that targeting LPCAT3 in adipose tissue to manipulate membrane phospholipid saturation is a new strategy to treat insulin resistance. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Mulin He
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Zhiqiang Li
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Victoria Sook Keng Tung
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Meixia Pan
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Xianlin Han
- Lipidomics Core, The University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Oleg Evgrafov
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
| | - Xian-Cheng Jiang
- Department of Cell Biology, The State University of New York Downstate Health Sciences University, Brooklyn, NY
- Molecular and Cellular Cardiology Program, Veterans Affairs New York Harbor Healthcare System, New York, NY
| |
Collapse
|
25
|
Zhao Q, Yu C, Xu X, Jin W, Zhang Z, Huang H, Gao Y, Pan D. Phosphorylated YBX2 is stabilized to promote glycolysis in brown adipocytes. iScience 2023; 26:108091. [PMID: 37860762 PMCID: PMC10583057 DOI: 10.1016/j.isci.2023.108091] [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: 05/05/2023] [Revised: 08/24/2023] [Accepted: 09/26/2023] [Indexed: 10/21/2023] Open
Abstract
Y-box binding protein 2 (YBX2) is an essential modulator of brown adipose tissue activation, yet the regulation on its own expression and the involved mechanism remains largely unknown. Herein, we report the YBX2 protein level, but not mRNA level, is induced in response to acute β-adrenergic signaling. In this context, YBX2 is a dual substrate for both AMPK and Akt2. The phosphorylation at Thr115 by AMPK or at Ser137 by Akt2 facilitates YBX2 accumulation in brown adipocytes by decreasing ubiquitination-mediated degradation. Beyond stabilizing PGC1α mRNA, increased YBX2 upon thermogenic activation assists the expression of glycolytic enzymes, promotes glucose utilization and lactate production. Mechanistically, YBX2 modulates translation of glycolytic genes via direct binding to 5'-UTRs of these genes. Together these findings suggest YBX2 is responsive to thermogenic stimuli by phosphorylation modification, and stabilized YBX2 helps to boost glycolysis and thermogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Qingwen Zhao
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Yu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xiaoxuan Xu
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Wenfang Jin
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Zhe Zhang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Haiyan Huang
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yue Gao
- Department of Geriatrics, Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Dongning Pan
- Key Laboratory of Metabolism and Molecular Medicine of the Ministry of Education, Department of Biochemistry and Molecular Biology of School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
26
|
U-Din M, de Mello VD, Tuomainen M, Raiko J, Niemi T, Fromme T, Klåvus A, Gautier N, Haimilahti K, Lehtonen M, Kristiansen K, Newman JW, Pietiläinen KH, Pihlajamäki J, Amri EZ, Klingenspor M, Nuutila P, Pirinen E, Hanhineva K, Virtanen KA. Cold-stimulated brown adipose tissue activation is related to changes in serum metabolites relevant to NAD + metabolism in humans. Cell Rep 2023; 42:113131. [PMID: 37708023 DOI: 10.1016/j.celrep.2023.113131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/06/2023] [Accepted: 08/29/2023] [Indexed: 09/16/2023] Open
Abstract
Cold-induced brown adipose tissue (BAT) activation is considered to improve metabolic health. In murine BAT, cold increases the fundamental molecule for mitochondrial function, nicotinamide adenine dinucleotide (NAD+), but limited knowledge of NAD+ metabolism during cold in human BAT metabolism exists. We show that cold increases the serum metabolites of the NAD+ salvage pathway (nicotinamide and 1-methylnicotinamide) in humans. Additionally, individuals with cold-stimulated BAT activation have decreased levels of metabolites from the de novo NAD+ biosynthesis pathway (tryptophan, kynurenine). Serum nicotinamide correlates positively with cold-stimulated BAT activation, whereas tryptophan and kynurenine correlate negatively. Furthermore, the expression of genes involved in NAD+ biosynthesis in BAT is related to markers of metabolic health. Our data indicate that cold increases serum tryptophan conversion to nicotinamide to be further utilized by BAT. We conclude that NAD+ metabolism is activated upon cold in humans and is probably regulated in a coordinated fashion by several tissues.
Collapse
Affiliation(s)
- Mueez U-Din
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland
| | - Vanessa D de Mello
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Marjo Tuomainen
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | - Juho Raiko
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Tarja Niemi
- Department of Surgery, Turku University Hospital, Turku, Finland
| | - Tobias Fromme
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Anton Klåvus
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland
| | | | - Kimmo Haimilahti
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Program for Stem Cells and Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Marko Lehtonen
- Department of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | | | - John W Newman
- Obesity and Metabolism Research Unit, USDA-ARS Western Human Nutrition Research Center, Davis, CA, USA; West Coast Metabolomics Center, Davis Genome Center, University of California, Davis, Davis, CA 95616, USA; Department of Nutrition, University of California, Davis, Davis, CA 95616, USA
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jussi Pihlajamäki
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | | | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich, Freising, Germany; EKFZ - Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany; ZIEL - Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Pirjo Nuutila
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Eija Pirinen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland; Research Unit for Internal Medicine, Faculty of Medicine, University of Oulu, 90220 Oulu, Finland
| | - Kati Hanhineva
- Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Life Technologies, Food Chemistry and Food Development Unit, University of Turku, Turku, Finland; Department of Biology and Biological Engineering, Division of Food and Nutrition Science, Chalmers University of Technology, Gothenburg, Sweden
| | - Kirsi A Virtanen
- Turku PET Centre, Turku University Hospital, Turku, Finland; Turku PET Centre, University of Turku, Turku, Finland; Department of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Department of Medicine, Kuopio University Hospital, Kuopio, Finland; Department of Endocrinology, Turku University Hospital, Turku, Finland.
| |
Collapse
|
27
|
Nikanorova AA, Barashkov NA, Pshennikova VG, Teryutin FM, Nakhodkin SS, Solovyev AV, Romanov GP, Burtseva TE, Fedorova SA. A Systematic Review and Meta-Analysis of Free Triiodothyronine (FT3) Levels in Humans Depending on Seasonal Air Temperature Changes: Is the Variation in FT3 Levels Related to Nonshivering Thermogenesis? Int J Mol Sci 2023; 24:14052. [PMID: 37762355 PMCID: PMC10531421 DOI: 10.3390/ijms241814052] [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: 08/22/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Thyroid hormones play a crucial role in regulating normal development, growth, and metabolic function. However, the controversy surrounding seasonal changes in free triiodothyronine (FT3) levels remains unresolved. Therefore, the aim of this study was to conduct a systematic review and meta-analysis of variations in FT3 levels in relation to seasonal air temperatures in the context of current knowledge about its role in nonshivering thermogenesis. Ten eligible articles with a total of 336,755 participants were included in the meta-analysis. The studies were categorized into two groups based on the air temperature: "Cold winter", where the winter temperature fell below 0 °C, and "Warm winter", where the winter temperature was above 0 °C. The analysis revealed that in cold regions, FT3 levels decreased in winter compared to summer (I2 = 57%, p < 0.001), whereas in warm regions, FT3 levels increased during winter (I2 = 28%, p < 0.001). These findings suggest that seasonal variations in FT3 levels are likely to be influenced by the winter temperature. Considering the important role of the FT3 in the nonshivering thermogenesis process, we assume that this observed pattern is probably related to the differences in use of thyroid hormones in the brown adipose tissue during adaptive thermogenesis, which may depend on intensity of cold exposure.
Collapse
Affiliation(s)
- Alena A. Nikanorova
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
| | - Nikolay A. Barashkov
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
| | - Vera G. Pshennikova
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
| | - Fedor M. Teryutin
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
| | - Sergey S. Nakhodkin
- M.K. Ammosov North-Eastern Federal University, Kulakovskogo 46, 677013 Yakutsk, Russia; (S.S.N.); (A.V.S.); (G.P.R.)
| | - Aisen V. Solovyev
- M.K. Ammosov North-Eastern Federal University, Kulakovskogo 46, 677013 Yakutsk, Russia; (S.S.N.); (A.V.S.); (G.P.R.)
| | - Georgii P. Romanov
- M.K. Ammosov North-Eastern Federal University, Kulakovskogo 46, 677013 Yakutsk, Russia; (S.S.N.); (A.V.S.); (G.P.R.)
| | - Tatiana E. Burtseva
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
- M.K. Ammosov North-Eastern Federal University, Kulakovskogo 46, 677013 Yakutsk, Russia; (S.S.N.); (A.V.S.); (G.P.R.)
| | - Sardana A. Fedorova
- Yakut Science Centre of Complex Medical Problems, Yaroslavskogo 6/3, 677000 Yakutsk, Russia; (A.A.N.); (V.G.P.); (F.M.T.); (T.E.B.); (S.A.F.)
- M.K. Ammosov North-Eastern Federal University, Kulakovskogo 46, 677013 Yakutsk, Russia; (S.S.N.); (A.V.S.); (G.P.R.)
| |
Collapse
|
28
|
Xu J, Cui L, Wang J, Zheng S, Zhang H, Ke S, Cao X, Shi Y, Li J, Zen K, Vidal-Puig A, Zhang CY, Li L, Jiang X. Cold-activated brown fat-derived extracellular vesicle-miR-378a-3p stimulates hepatic gluconeogenesis in male mice. Nat Commun 2023; 14:5480. [PMID: 37673898 PMCID: PMC10482845 DOI: 10.1038/s41467-023-41160-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 08/24/2023] [Indexed: 09/08/2023] Open
Abstract
During cold exposure, activated brown adipose tissue (BAT) takes up a large amount of circulating glucose to fuel non-shivering thermogenesis and defend against hypothermia. However, little is known about the endocrine function of BAT controlling glucose homoeostasis under this thermoregulatory challenge. Here, we show that in male mice, activated BAT-derived extracellular vesicles (BDEVs) reprogram systemic glucose metabolism by promoting hepatic gluconeogenesis during cold stress. Cold exposure facilitates the selective packaging of miR-378a-3p-one of the BAT-enriched miRNAs-into EVs and delivery into the liver. BAT-derived miR-378a-3p enhances gluconeogenesis by targeting p110α. miR-378 KO mice display reduced hepatic gluconeogenesis during cold exposure, while restoration of miR-378a-3p in iBAT induces the expression of gluconeogenic genes in the liver. These findings provide a mechanistic understanding of BDEV-miRNA as stress-induced batokine to coordinate systemic glucose homoeostasis. This miR-378a-3p-mediated interorgan communication highlights a novel endocrine function of BAT in preventing hypoglycemia during cold stress.
Collapse
Affiliation(s)
- Jinhong Xu
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Le Cui
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Jiaqi Wang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Shasha Zheng
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Huahua Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Shuo Ke
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Xiaoqin Cao
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Yanteng Shi
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Jing Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Ke Zen
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge Metabolic Research Laboratories, Cambridge, UK.
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, China.
| | - Chen-Yu Zhang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, China.
- Research Unit of Extracellular RNA, Chinese Academy of Medical Sciences, Nanjing, Jiangsu, China.
- Institute of Artificial Intelligence Biomedicine, Nanjing University, Nanjing, Jiangsu, China.
| | - Liang Li
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, China.
| | - Xiaohong Jiang
- Nanjing Drum Tower Hospital Center of Molecular Diagnostic and Therapy, State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, Jiangsu, China.
- Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, China.
- Jiangsu Engineering Research Center for MicroRNA Biology and Biotechnology, NJU Advanced Institute of Life Sciences (NAILS), Nanjing, Jiangsu, China.
| |
Collapse
|
29
|
Liu Y, Qu Y, Cheng C, Tsai PY, Edwards K, Xue S, Pandit S, Eguchi S, Sanghera N, Barrow JJ. Nipsnap1-A regulatory factor required for long-term maintenance of non-shivering thermogenesis. Mol Metab 2023; 75:101770. [PMID: 37423391 PMCID: PMC10404556 DOI: 10.1016/j.molmet.2023.101770] [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: 02/16/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/11/2023] Open
Abstract
OBJECTIVE The activation of non-shivering thermogenesis (NST) has strong potential to combat obesity and metabolic disease. The activation of NST however is extremely temporal and the mechanisms surrounding how the benefits of NST are sustained once fully activated, remain unexplored. The objective of this study is to investigate the role of 4-Nitrophenylphosphatase Domain and Non-Neuronal SNAP25-Like 1 (Nipsnap1) in NST maintenance, which is a critical regulator identified in this study. METHODS The expression of Nipsnap1 was profiled by immunoblotting and RT-qPCR. We generated Nipsnap1 knockout mice (N1-KO) and investigated the function of Nipsnap1 in NST maintenance and whole-body metabolism using whole body respirometry analyses. We evaluate the metabolic regulatory role of Nipsnap1 using cellular and mitochondrial respiration assay. RESULTS Here, we show Nipsnap1 as a critical regulator of long-term thermogenic maintenance in brown adipose tissue (BAT). Nipsnap1 localizes to the mitochondrial matrix and increases its transcript and protein levels in response to both chronic cold and β3 adrenergic signaling. We demonstrated that these mice are unable to sustain activated energy expenditure and have significantly lower body temperature in the face of an extended cold challenge. Furthermore, when mice are exposed to the pharmacological β3 agonist CL 316, 243, the N1-KO mice exhibit significant hyperphagia and altered energy balance. Mechanistically, we demonstrate that Nipsnap1 integrates with lipid metabolism and BAT-specific ablation of Nipsnap1 leads to severe defects in beta-oxidation capacity when exposed to a cold environmental challenge. CONCLUSION Our findings identify Nipsnap1 as a potent regulator of long-term NST maintenance in BAT.
Collapse
Affiliation(s)
- Yang Liu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Yue Qu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Chloe Cheng
- Department of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Pei-Yin Tsai
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Kaydine Edwards
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Siwen Xue
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Supriya Pandit
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Sakura Eguchi
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA
| | - Navneet Sanghera
- Department of Biological Sciences, San Jose State University, San Jose, CA, 95192, USA
| | - Joeva J Barrow
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
30
|
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
|
31
|
Gong L, Zhao S, Chu X, Yang H, Li Y, Wei S, Li F, Zhang Y, Li S, Jiang P. Assessment of cold exposure-induced metabolic changes in mice using untargeted metabolomics. Front Mol Biosci 2023; 10:1228771. [PMID: 37719264 PMCID: PMC10500074 DOI: 10.3389/fmolb.2023.1228771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/18/2023] [Indexed: 09/19/2023] Open
Abstract
Background: Cold exposure (CE) can effectively modulate adipose tissue metabolism and improve metabolic health. Although previous metabolomics studies have primarily focused on analyzing one or two samples from serum, brown adipose tissue (BAT), white adipose tissue (WAT), and liver samples, there is a significant lack of simultaneous analysis of multiple tissues regarding the metabolic changes induced by CE in mice. Therefore, our study aims to investigate the metabolic profiles of the major tissues involved. Methods: A total of 14 male C57BL/6J mice were randomly assigned to two groups: the control group (n = 7) and the CE group (n = 7). Metabolite determination was carried out using gas chromatography-mass spectrometry (GC-MS), and multivariate analysis was employed to identify metabolites exhibiting differential expression between the two groups. Results: In our study, we identified 32 discriminant metabolites in BAT, 17 in WAT, 21 in serum, 7 in the liver, 16 in the spleen, and 26 in the kidney, respectively. Among these metabolites, amino acids, fatty acids, and nucleotides emerged as the most significantly altered compounds. These metabolites were found to be associated with 12 differential metabolic pathways closely related to amino acids, fatty acids, and energy metabolism. Conclusion: Our study may provide valuable insights into the metabolic effects induced by CE, and they have the potential to inspire novel approaches for treating metabolic diseases.
Collapse
Affiliation(s)
| | - Shiyuan Zhao
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| | - Xue Chu
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
| | - Hui Yang
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yanan Li
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Shanshan Wei
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Graduate Department, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, China
| | - Fengfeng Li
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Yazhou Zhang
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Shuhui Li
- Tengzhou Central People’s Hospital, Tengzhou, China
| | - Pei Jiang
- Translational Pharmaceutical Laboratory, Jining First People’s Hospital, Shandong First Medical University, Jining, China
- Institute of Translational Pharmacy, Jining Medical Research Academy, Jining, China
| |
Collapse
|
32
|
Laiglesia LM, Escoté X, Sáinz N, Felix-Soriano E, Santamaría E, Collantes M, Fernández-Galilea M, Colón-Mesa I, Martínez-Fernández L, Quesada-López T, Quesada-Vázquez S, Rodríguez-Ortigosa C, Arbones-Mainar JM, Valverde ÁM, Martínez JA, Dalli J, Herrero L, Lorente-Cebrián S, Villarroya F, Moreno-Aliaga MJ. Maresin 1 activates brown adipose tissue and promotes browning of white adipose tissue in mice. Mol Metab 2023; 74:101749. [PMID: 37271337 PMCID: PMC10331312 DOI: 10.1016/j.molmet.2023.101749] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 05/19/2023] [Accepted: 06/01/2023] [Indexed: 06/06/2023] Open
Abstract
OBJECTIVE Maresin 1 (MaR1) is a docosahexaenoic acid-derived proresolving lipid mediator with insulin-sensitizing and anti-steatosis properties. Here, we aim to unravel MaR1 actions on brown adipose tissue (BAT) activation and white adipose tissue (WAT) browning. METHODS MaR1 actions were tested in cultured murine brown adipocytes and in human mesenchymal stem cells (hMSC)-derived adipocytes. In vivo effects of MaR1 were tested in diet-induced obese (DIO) mice and lean WT and Il6 knockout (Il6-/-) mice. RESULTS In cultured differentiated murine brown adipocytes, MaR1 reduces the expression of inflammatory genes, while stimulates glucose uptake, fatty acid utilization and oxygen consumption rate, along with the upregulation of mitochondrial mass and genes involved in mitochondrial biogenesis and function and the thermogenic program. In Leucine Rich Repeat Containing G Protein-Coupled Receptor 6 (LGR6)-depleted brown adipocytes using siRNA, the stimulatory effect of MaR1 on thermogenic genes was abrogated. In DIO mice, MaR1 promotes BAT remodeling, characterized by higher expression of genes encoding for master regulators of mitochondrial biogenesis and function and iBAT thermogenic activation, together with increased M2 macrophage markers. In addition, MaR1-treated DIO mice exhibit a better response to cold-induced BAT activation. Moreover, MaR1 induces a beige adipocyte signature in inguinal WAT of DIO mice and in hMSC-derived adipocytes. MaR1 potentiates Il6 expression in brown adipocytes and BAT of cold exposed lean WT mice. Interestingly, the thermogenic properties of MaR1 were abrogated in Il6-/- mice. CONCLUSIONS These data reveal MaR1 as a novel agent that promotes BAT activation and WAT browning by regulating thermogenic program in adipocytes and M2 polarization of macrophages. Moreover, our data suggest that LGR6 receptor is mediating MaR1 actions on brown adipocytes, and that IL-6 is required for the thermogenic effects of MaR1.
Collapse
Affiliation(s)
- Laura M Laiglesia
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Xavier Escoté
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Reus, 43204 Spain
| | - Neira Sáinz
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Elisa Felix-Soriano
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Eva Santamaría
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), Instituto de Salud Carlos III (ISCIII), Madrid 28029, Spain; Division of Hepatology, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - María Collantes
- Department of Nuclear Medicine/ Translational Molecular Imaging Unit (UNIMTRA), Clínica Universidad de Navarra, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Marta Fernández-Galilea
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Ignacio Colón-Mesa
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Leyre Martínez-Fernández
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain
| | - Tania Quesada-López
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | | | | | - José M Arbones-Mainar
- Adipocyte and Fat Biology Laboratory (AdipoFat), Instituto de Investigación Sanitaria Aragón, Instituto Aragonés de Ciencias de la Salud, Unidad de Investigación Traslacional, Hospital Universitario Miguel Servet, Zaragoza, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Ángela M Valverde
- Alberto Sols Biomedical Research Institute (IIBm) (CSIC/UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - J Alfredo Martínez
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK; Center for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Laura Herrero
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain; Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, Institut de Biomedicina de la Universitat de Barcelona (IBUB), Universitat de Barcelona, Barcelona, Spain
| | - Silvia Lorente-Cebrián
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Current address: Department of Pharmacology, Physiology, Legal and Forensic Medicine. Faculty of Health and Sport Science, University of Zaragoza, Zaragoza, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Institute of Biomedicine of the University of Barcelona, Barcelona, Catalonia, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - María J Moreno-Aliaga
- University of Navarra, Center for Nutrition Research, Pamplona, 31008, Spain; University of Navarra, Department of Nutrition, Food Science and Physiology, Pamplona, 31008, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
| |
Collapse
|
33
|
Maliszewska K, Miniewska K, Godlewski A, Gosk W, Mojsak M, Kretowski A, Ciborowski M. Changes in plasma endocannabinoids concentrations correlate with 18F-FDG PET/MR uptake in brown adipocytes in humans. Front Mol Biosci 2023; 10:1073683. [PMID: 37564131 PMCID: PMC10411954 DOI: 10.3389/fmolb.2023.1073683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 07/13/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction: Recent data suggest a possible role of endocannabinoids in the regulation of brown adipose tissue (BAT) activity. Those findings indicate potential treatment options for obesity. The aim of this study was to evaluate the relationship between plasma endocannabinoids concentrations and the presence of BAT in humans. Methods: The study group consisted of 25 subjects divided into two groups: BAT positive BAT(+), (n = 17, median age = 25 years) and BAT negative BAT(-), (n = 8, median age = 28 years). BAT was estimated using 18F-FDG PET/MR after 2 h of cold exposure. The level of plasma endocannabinoids was assessed at baseline, 60 min and 120 min of cold exposure. Results: In both groups, BAT(+) and BAT(-), during the cooling, we observed a decrease of the same endocannabinoids: arachidonoylethanolamide (AEA), eicosapentaenoyl ethanolamide (EPEA) and oleoyl ethanolamide (OEA) with a much more profound decline in BAT(+) subjects. Statistically significant fall of PEA (palmitoylethanolamide) and SEA (stearoylethanolamide) concentrations after 60 min (FC = 0.7, p = 0.007 and FC = 0.8, p = 0.03, respectively) and 120 min (FC = 0.81, p = 0.004, and FC = 0.9, p = 0.01, respectively) of cooling was observed only in individuals with BAT. Conclusion: We noticed the profound decline of endocannabinoids concentrations in subjects with increased 18F-FDG PET/MR uptake in BAT. Identification of a new molecules related to BAT activity may create a new target for obesity treatment.
Collapse
Affiliation(s)
- Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Katarzyna Miniewska
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Adrian Godlewski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Wioleta Gosk
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| | - Malgorzata Mojsak
- Independent Laboratory of Molecular Imaging, Medical University of Bialystok, Bialystok, Poland
| | - Adam Kretowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Bialystok, Poland
| | - Michal Ciborowski
- Clinical Research Centre, Medical University of Bialystok, Bialystok, Poland
| |
Collapse
|
34
|
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
|
35
|
Pellegrinelli V, Figueroa-Juárez E, Samuelson I, U-Din M, Rodriguez-Fdez S, Virtue S, Leggat J, Çubuk C, Peirce VJ, Niemi T, Campbell M, Rodriguez-Cuenca S, Blázquez JD, Carobbio S, Virtanen KA, Vidal-Puig A. Defective extracellular matrix remodeling in brown adipose tissue is associated with fibro-inflammation and reduced diet-induced thermogenesis. Cell Rep 2023; 42:112640. [PMID: 37318951 DOI: 10.1016/j.celrep.2023.112640] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/25/2023] [Accepted: 05/29/2023] [Indexed: 06/17/2023] Open
Abstract
The relevance of extracellular matrix (ECM) remodeling is reported in white adipose tissue (AT) and obesity-related dysfunctions, but little is known about the importance of ECM remodeling in brown AT (BAT) function. Here, we show that a time course of high-fat diet (HFD) feeding progressively impairs diet-induced thermogenesis concomitantly with the development of fibro-inflammation in BAT. Higher markers of fibro-inflammation are associated with lower cold-induced BAT activity in humans. Similarly, when mice are housed at thermoneutrality, inactivated BAT features fibro-inflammation. We validate the pathophysiological relevance of BAT ECM remodeling in response to temperature challenges and HFD using a model of a primary defect in the collagen turnover mediated by partial ablation of the Pepd prolidase. Pepd-heterozygous mice display exacerbated dysfunction and BAT fibro-inflammation at thermoneutrality and in HFD. Our findings show the relevance of ECM remodeling in BAT activation and provide a mechanism for BAT dysfunction in obesity.
Collapse
Affiliation(s)
- Vanessa Pellegrinelli
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
| | - Elizabeth Figueroa-Juárez
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Isabella Samuelson
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Mueez U-Din
- Turku PET Centre, University of Turku, Turku, Finland; Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Sonia Rodriguez-Fdez
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Samuel Virtue
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Jennifer Leggat
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Cankut Çubuk
- Platform of Computational Medicine, Fundación Progreso y Salud (FPS), Hospital Virgen Del Rocío, 41013 Sevilla, Spain
| | - Vivian J Peirce
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Tarja Niemi
- Department of Plastic and General Surgery, Turku University Hospital, Turku, Finland
| | - Mark Campbell
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P.R. China
| | - Sergio Rodriguez-Cuenca
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P.R. China
| | - Joaquin Dopazo Blázquez
- Platform of Computational Medicine, Fundación Progreso y Salud (FPS), Hospital Virgen Del Rocío, 41013 Sevilla, Spain; Bioinformatics in RareDiseases (BiER), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), 41013 Sevilla, Spain; Computational Systems Medicine, Institute of Biomedicine of Seville (IBiS), Sevilla 41013, Spain; Functional Genomics Node (INB-ELIXIR-es), Sevilla, Spain
| | - Stefania Carobbio
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain
| | - Kirsi A Virtanen
- Turku PET Centre, University of Turku, Turku, Finland; Institute of Public Health and Clinical Nutrition, University of Eastern Finland (UEF), Kuopio, Finland; Department of Endocrinology and Clinical Nutrition, Kuopio University Hospital, Kuopio, Finland
| | - Antonio Vidal-Puig
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, P.R. China; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain; Cambridge Heart and Lung Research Institute, Cambridge, UK.
| |
Collapse
|
36
|
Blondin DP. Human thermogenic adipose tissue. Curr Opin Genet Dev 2023; 80:102054. [PMID: 37269791 DOI: 10.1016/j.gde.2023.102054] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 04/11/2023] [Accepted: 04/28/2023] [Indexed: 06/05/2023]
Abstract
Human thermogenic adipose tissue has long been touted as a promising therapeutic target for obesity and its associated metabolic diseases. Here, we provide a brief overview of the current knowledge of in vivo human thermogenic adipose tissue metabolism. We explore the evidence provided by retrospective and prospective studies describing the association of brown adipose tissue (BAT) [18F]fluorodeoxyglucose accumulation and various cardiometabolic risk factors. Although these studies have been invaluable in generating hypothesis, it has also raised some questions about the reliability of this method as an indicator of BAT thermogenic capacity. We discuss the evidence in support of human BAT functioning as a local thermogenic organ and energy sink, as an endocrine organ, and as a biomarker of adipose tissue health.
Collapse
Affiliation(s)
- Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, 3001, 12th Ave North, Sherbrooke, Quebec J1H 5N4, Canada.
| |
Collapse
|
37
|
Graceli JB, da Costa CS, Laws MJ, Deviney ARK, Meling D, Flaws JA. Chronic exposure to a mixture of phthalates shifts the white and brown adipose tissue phenotypes in female mice. Toxicol Sci 2023; 193:204-218. [PMID: 37021957 PMCID: PMC10230284 DOI: 10.1093/toxsci/kfad032] [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] [Indexed: 04/07/2023] Open
Abstract
Phthalates are endocrine-disrupting chemicals used in consumer products. Although phthalates are obesogens and affect metabolic function, it is unknown if chronic exposure for 6 months to a phthalate mixture alters adipose tissue phenotype in female mice. After vehicle or mixture exposure, white adipose tissue and brown adipose tissue (WAT and BAT) were analyzed for expression of adipogenesis, proliferation, angiogenesis, apoptosis, oxidative stress, inflammation, and collagen deposition markers. The mixture altered WAT morphology, leading to an increase in hyperplasia, blood vessel number, and expression of BAT markers (Adipoq and Fgf2) in WAT. The mixture increased the expression of the inflammatory markers, Il1β, Ccl2, and Ccl5, in WAT. The mixture also increased expression of the proapoptotic (Bax and Bcl2) and antiapoptotic (Bcl2l10) factors in WAT. The mixture increased expression of the antioxidant Gpx1 in WAT. The mixture changed BAT morphology by increasing adipocyte diameter, whitening area, and blood vessel number and decreased expression of the thermogenic markers Ucp1, Pgargc1a, and Adrb3. Furthermore, the mixture increased the expression of adipogenic markers Plin1 and Cebpa, increased mast cell number, and increased Il1β expression in BAT. The mixture also increased expression of the antioxidant markers Gpx and Nrf2 and the apoptotic marker Casp2 in BAT. Collectively, these data indicate that chronic exposure to a phthalate mixture alters WAT and BAT lipid metabolism phenotypes in female mice, leading to an apparent shift in their normal morphology. Following long-term exposure to a phthalate mixture, WAT presented BAT-like features and BAT presented WAT-like features.
Collapse
Affiliation(s)
- Jones B Graceli
- Department of Morphology, Federal University of Espirito Santo, Vitoria, Brazil
| | - Charles S da Costa
- Department of Morphology, Federal University of Espirito Santo, Vitoria, Brazil
| | - Mary J Laws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Ashley R K Deviney
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Daryl Meling
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Jodi A Flaws
- Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
38
|
Hropot T, Herman R, Janez A, Lezaic L, Jensterle M. Brown Adipose Tissue: A New Potential Target for Glucagon-like Peptide 1 Receptor Agonists in the Treatment of Obesity. Int J Mol Sci 2023; 24:ijms24108592. [PMID: 37239935 DOI: 10.3390/ijms24108592] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 05/01/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Adipose tissue can be divided into white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue, according to the differences in morphology. WAT acts as a buffer for increased energy intake and decreased energy expenditure during the development of obesity, resulting in visceral and ectopic WAT accumulation. These WAT depots are strongly associated with chronic systemic inflammation, insulin resistance, and cardiometabolic risk related to obesity. They represent a primary weight loss target in anti-obesity management. Second-generation anti-obesity medications glucagon-like peptide-1 receptor agonists (GLP-1RAs) cause weight loss and improve body composition by reducing visceral and ectopic fat depots of WAT, resulting in improved cardiometabolic health. Recently, the understanding of the physiological significance of BAT beyond its primary function in generating heat through non-shivering thermogenesis has been expanded. This has raised scientific and pharmaceutical interest in the manipulation of BAT to further enhance weight reduction and body weight maintenance. This narrative review focuses on the potential impact of GLP-1 receptor agonism on BAT, particularly in human clinical studies. It provides an overview of the role of BAT in weight management and highlights the need for further research to elucidate the mechanisms by which GLP-1RAs affect energy metabolism and weight loss. Despite encouraging preclinical data, limited clinical evidence supports the notion that GLP-1RAs contribute to BAT activation.
Collapse
Affiliation(s)
- Tim Hropot
- Department of Pediatric Endocrinology, Diabetes and Metabolic Diseases, University Children's Hospital, 1000 Ljubljana, Slovenia
| | - Rok Herman
- Department for Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Andrej Janez
- Department for Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Luka Lezaic
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department for Nuclear Medicine, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
| | - Mojca Jensterle
- Department for Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre Ljubljana, 1000 Ljubljana, Slovenia
- Department of Internal Medicine, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| |
Collapse
|
39
|
Sun L, Laurila S, Lahesmaa M, Rebelos E, Virtanen KA, Schnabl K, Klingenspor M, Nummenmaa L, Nuutila P. Secretin modulates appetite via brown adipose tissue-brain axis. Eur J Nucl Med Mol Imaging 2023; 50:1597-1606. [PMID: 36764966 PMCID: PMC10119257 DOI: 10.1007/s00259-023-06124-4] [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: 09/08/2022] [Accepted: 01/21/2023] [Indexed: 02/12/2023]
Abstract
PURPOSE Secretin activates brown adipose tissue (BAT) and induces satiation in both mice and humans. However, the exact brain mechanism of this satiety inducing, secretin-mediated gut-BAT-brain axis is largely unknown. METHODS AND RESULTS In this placebo-controlled, single-blinded neuroimaging study, firstly using [18F]-fluorodeoxyglucose (FDG) PET measures (n = 15), we established that secretin modulated brain glucose consumption through the BAT-brain axis. Predominantly, we found that BAT and caudate glucose uptake levels were negatively correlated (r = -0.54, p = 0.037) during secretin but not placebo condition. Then, using functional magnetic resonance imaging (fMRI; n = 14), we found that secretin improved inhibitory control and downregulated the brain response to appetizing food images. Finally, in a PET-fMRI fusion analysis (n = 10), we disclosed the patterned correspondence between caudate glucose uptake and neuroactivity to reward and inhibition, showing that the secretin-induced neurometabolic coupling patterns promoted satiation. CONCLUSION These findings suggest that secretin may modulate the BAT-brain metabolic crosstalk and subsequently the neurometabolic coupling to induce satiation. The study advances our understanding of the secretin signaling in motivated eating behavior and highlights the potential role of secretin in treating eating disorders and obesity. TRIAL REGISTRATION EudraCT no. 2016-002373-35, registered 2 June 2016; Clinical Trials no. NCT03290846, registered 25 September 2017.
Collapse
Affiliation(s)
- Lihua Sun
- Department of Nuclear Medicine, Pudong Hospital, Fudan University, Shanghai, China.
- Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, China.
- Turku PET Centre, University of Turku, Turku, Finland.
- Turku PET Centre, Turku University Hospital, Turku, Finland.
| | - Sanna Laurila
- Turku PET Centre, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
- Department of Medicine, University of Turku, Turku, Finland
| | - Minna Lahesmaa
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Eleni Rebelos
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Kirsi A Virtanen
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Katharina Schnabl
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich, TUM School of Life Sciences, Freising, Germany
- EKFZ-Else Kröner Fresenius Center for Nutritional Medicine, Technical University of Munich, Freising, Germany
- ZIEL-Institute for Food & Health, Technical University of Munich, Freising, Germany
| | - Lauri Nummenmaa
- Turku PET Centre, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
- Department of Psychology, University of Turku, Turku, Finland
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
40
|
Della Guardia L, Wang L. Fine particulate matter induces adipose tissue expansion and weight gain: Pathophysiology. Obes Rev 2023; 24:e13552. [PMID: 36700515 DOI: 10.1111/obr.13552] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/25/2022] [Accepted: 01/08/2023] [Indexed: 01/27/2023]
Abstract
Dysregulations in energy balance represent a major driver of obesity. Recent evidence suggests that environmental factors also play a pivotal role in inducing weight gain. Chronic exposure to fine particulate matter (PM2.5 ) is associated with white adipose tissue (WAT) expansion in animals and higher rates of obesity in humans. This review discusses metabolic adaptions in central and peripheral tissues that promote energy storage and WAT accumulation in PM2.5 -exposed animals and humans. Chronic PM2.5 exposure produces inflammation and leptin resistance in the hypothalamus, decreasing energy expenditure and increasing food intake. PM2.5 promotes the conversion of brown adipocytes toward the white phenotype, resulting in decreased energy expenditure. The development of inflammation in WAT can stimulate adipogenesis and hampers catecholamine-induced lipolysis. PM2.5 exposure affects the thyroid, reducing the release of thyroxine and tetraiodothyronine. In addition, PM2.5 exposure compromises skeletal muscle fitness by inhibiting Nitric oxide (NO)-dependent microvessel dilation and impairing mitochondrial oxidative capacity, with negative effects on energy expenditure. This evidence suggests that pathological alterations in the hypothalamus, brown adipose tissue, WAT, thyroid, and skeletal muscle can alter energy homeostasis, increasing lipid storage and weight gain in PM2.5 -exposed animals and humans. Further studies will enrich this pathophysiological model.
Collapse
Affiliation(s)
- Lucio Della Guardia
- Department of Biomedical Sciences for Health, Università degli Studi di Milano, Milan, Italy
| | - Ling Wang
- Hubei Key Laboratory of Environmental and Health Effects of Persistent Toxic Substances, School of Environment and Health, Jianghan University, Wuhan, China
| |
Collapse
|
41
|
Thermogenic Capacity of Human Supraclavicular Brown Fat and Cold-Stimulated Brain Glucose Metabolism. Metabolites 2023; 13:metabo13030387. [PMID: 36984827 PMCID: PMC10055954 DOI: 10.3390/metabo13030387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Human brain metabolism is susceptible to temperature changes. It has been suggested that the supraclavicular brown adipose tissue (BAT) protects the brain from these fluctuations by regulating heat production through the presence of uncoupling protein 1 (UCP-1). It remains unsolved whether inter-individual variation in the expression of UCP-1, which represents the thermogenic capacity of the supraclavicular BAT, is linked with brain metabolism during cold stress. Ten healthy human participants underwent 18F-FDG PET scanning of the brain under cold stimulus to determine brain glucose uptake (BGU). On a separate day, an excision biopsy of the supraclavicular fat—the fat proximal to the carotid arteries supplying the brain with warm blood—was performed to determine the mRNA expression of the thermogenic protein UCP-1. Expression of UCP-1 in supraclavicular BAT was directly related to the whole brain glucose uptake rate determined under cold stimulation (rho = 0.71, p = 0.03). In sub-compartmental brain analysis, UCP-1 expression in supraclavicular BAT was directly related to cold-stimulated glucose uptake rates in the hypothalamus, medulla, midbrain, limbic system, frontal lobe, occipital lobe, and parietal lobe (all rho ≥ 0.67, p < 0.05). These relationships were independent of body mass index and age. When analysing gene expressions of BAT secretome, we found a positive correlation between cold-stimulated BGU and DIO2. These findings provide evidence of functional links between brain metabolism under cold stimulation and UCP-1 and DIO2 expressions in BAT in humans. More research is needed to evaluate the importance of these findings in clinical outcomes, for instance, in examining the supporting role of BAT in cognitive functions under cold stress.
Collapse
|
42
|
Abstract
Brown adipose tissue (BAT) displays the unique capacity to generate heat through uncoupled oxidative phosphorylation that makes it a very attractive therapeutic target for cardiometabolic diseases. Here, we review BAT cellular metabolism, its regulation by the central nervous and endocrine systems and circulating metabolites, the plausible roles of this tissue in human thermoregulation, energy balance, and cardiometabolic disorders, and the current knowledge on its pharmacological stimulation in humans. The current definition and measurement of BAT in human studies relies almost exclusively on BAT glucose uptake from positron emission tomography with 18F-fluorodeoxiglucose, which can be dissociated from BAT thermogenic activity, as for example in insulin-resistant states. The most important energy substrate for BAT thermogenesis is its intracellular fatty acid content mobilized from sympathetic stimulation of intracellular triglyceride lipolysis. This lipolytic BAT response is intertwined with that of white adipose (WAT) and other metabolic tissues, and cannot be independently stimulated with the drugs tested thus far. BAT is an interesting and biologically plausible target that has yet to be fully and selectively activated to increase the body's thermogenic response and shift energy balance. The field of human BAT research is in need of methods able to directly, specifically, and reliably measure BAT thermogenic capacity while also tracking the related thermogenic responses in WAT and other tissues. Until this is achieved, uncertainty will remain about the role played by this fascinating tissue in human cardiometabolic diseases.
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, Quebec, J1H 5N4, Canada
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, Quebec, J1H 5N4, Canada
| | | | - Denis Richard
- Centre de recherche de l’Institut universitaire de cardiologie et de pneumologie de Québec, Université Laval, Quebec City, Quebec, G1V 4G5, Canada
| |
Collapse
|
43
|
Merlo E, Zimerman J, Dos Santos FCF, Zanol JF, da Costa CS, Carneiro PH, Miranda-Alves L, Warner GR, Graceli JB. Subacute and low dose of tributyltin exposure leads to brown adipose abnormalities in male rats. Toxicol Lett 2023; 376:26-38. [PMID: 36638932 PMCID: PMC9928871 DOI: 10.1016/j.toxlet.2023.01.003] [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: 05/16/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Tributyltin (TBT) is an obesogenic endocrine disrupting chemical (EDC) linked with several metabolic complications. Brown adipose tissue (BAT) is the principal site for thermogenesis, making it a potential target for obesity management and metabolic disease. However, few studies have evaluated TBT effect on BAT function. In this investigation, we assessed whether subacute (15 days) and low dose of TBT exposure (100 ng/kg/day) results in abnormal BAT morphophysiology in adult male rats. Body temperature, BAT morphology, inflammation, oxidative stress, collagen deposition and BAT metabolic gene expression markers were assessed in room temperature (Room, ∼24 ºC) and after cold tolerance test (Cold, ∼4 ºC) conditions. A reduction in body temperature was observed in both Room and Cold conditions in TBT rats, suggesting abnormal BAT thermogenic function. Changes in BAT morphology were observed in TBT rats, with an increase in BAT lipid accumulation, an increase in BAT unilocular adipocyte number and a decrease in BAT multilocular adipocyte number in Room condition. All these parameters were opposite in Cold condition TBT rats, leading to a borderline increase in BAT UCP1 protein expression. An increase in BAT mast cell number was observed in TBT rats in Room condition. An increase in ED1 protein expression (macrophage marker) was observed in TBT rats in Cold condition. Oxidative stress and collagen deposition increased in both Room and Cold conditions in TBT rats. TBT exposure caused a borderline increase in BAT COL1A1 protein expression in Cold condition. Further, strong negative correlations were observed between body temperature and BAT lipid accumulation, and BAT lipid accumulation and multilocular adipocyte number. Thus, these data suggest that TBT exposure impaired BAT morphophysiology through impacts on lipid accumulation, inflammation, fibrosis and oxidative stress in male rats.
Collapse
Affiliation(s)
- Eduardo Merlo
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Jeanini Zimerman
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | | | - Jordana F Zanol
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Charles S da Costa
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil
| | - Pedro H Carneiro
- Experimental Endocrinology Research, Development and Innovation Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Postgraduate Program in Endocrinology, School of Medicine, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, Ilha do Governador, Cidade Universitária, UFRJ, RJ, Brazil
| | - Leandro Miranda-Alves
- Experimental Endocrinology Research, Development and Innovation Group, Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Brazil; Postgraduate Program in Endocrinology, School of Medicine, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho, Ilha do Governador, Cidade Universitária, UFRJ, RJ, Brazil
| | - Genoa R Warner
- Department of Chemistry and Environmental Science, New Jersey Institute of Technology, USA
| | - Jones B Graceli
- Department of Morphology, Federal University of Espírito Santo, Vitória, Brazil.
| |
Collapse
|
44
|
Straat ME, Hoekx CA, van Velden FHP, Pereira Arias-Bouda LM, Dumont L, Blondin DP, Boon MR, Martinez-Tellez B, Rensen PCN. Stimulation of the beta-2-adrenergic receptor with salbutamol activates human brown adipose tissue. Cell Rep Med 2023; 4:100942. [PMID: 36812890 PMCID: PMC9975328 DOI: 10.1016/j.xcrm.2023.100942] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/30/2022] [Accepted: 01/20/2023] [Indexed: 02/23/2023]
Abstract
While brown adipose tissue (BAT) is activated by the beta-3-adrenergic receptor (ADRB3) in rodents, in human brown adipocytes, the ADRB2 is dominantly present and responsible for noradrenergic activation. Therefore, we performed a randomized double-blinded crossover trial in young lean men to compare the effects of single intravenous bolus of the ADRB2 agonist salbutamol without and with the ADRB1/2 antagonist propranolol on glucose uptake by BAT, assessed by dynamic 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography-computed tomography scan (i.e., primary outcome). Salbutamol, compared with salbutamol with propranolol, increases glucose uptake by BAT, without affecting the glucose uptake by skeletal muscle and white adipose tissue. The salbutamol-induced glucose uptake by BAT positively associates with the increase in energy expenditure. Notably, participants with high salbutamol-induced glucose uptake by BAT have lower body fat mass, waist-hip ratio, and serum LDL-cholesterol concentration. In conclusion, specific ADRB2 agonism activates human BAT, which warrants investigation of ADRB2 activation in long-term studies (EudraCT: 2020-004059-34).
Collapse
Affiliation(s)
- Maaike E Straat
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Carlijn A Hoekx
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Floris H P van Velden
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lenka M Pereira Arias-Bouda
- Section of Nuclear Medicine, Department of Radiology, Leiden University Medical Center, 2333 ZA Leiden, the Netherlands
| | - Lauralyne Dumont
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Department of Physiology-Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Denis P Blondin
- Centre de Recherche du Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada; Department of Medicine, Division of Neurology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada
| | - Mariëtte R Boon
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Borja Martinez-Tellez
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, the Netherlands.
| |
Collapse
|
45
|
Ziqubu K, Dludla PV, Mthembu SXH, Nkambule BB, Mabhida SE, Jack BU, Nyambuya TM, Mazibuko-Mbeje SE. An insight into brown/beige adipose tissue whitening, a metabolic complication of obesity with the multifactorial origin. Front Endocrinol (Lausanne) 2023; 14:1114767. [PMID: 36875450 PMCID: PMC9978510 DOI: 10.3389/fendo.2023.1114767] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 02/06/2023] [Indexed: 02/18/2023] Open
Abstract
Brown adipose tissue (BAT), a thermoregulatory organ known to promote energy expenditure, has been extensively studied as a potential avenue to combat obesity. Although BAT is the opposite of white adipose tissue (WAT) which is responsible for energy storage, BAT shares thermogenic capacity with beige adipose tissue that emerges from WAT depots. This is unsurprising as both BAT and beige adipose tissue display a huge difference from WAT in terms of their secretory profile and physiological role. In obesity, the content of BAT and beige adipose tissue declines as these tissues acquire the WAT characteristics via the process called "whitening". This process has been rarely explored for its implication in obesity, whether it contributes to or exacerbates obesity. Emerging research has demonstrated that BAT/beige adipose tissue whitening is a sophisticated metabolic complication of obesity that is linked to multiple factors. The current review provides clarification on the influence of various factors such as diet, age, genetics, thermoneutrality, and chemical exposure on BAT/beige adipose tissue whitening. Moreover, the defects and mechanisms that underpin the whitening are described. Notably, the BAT/beige adipose tissue whitening can be marked by the accumulation of large unilocular lipid droplets, mitochondrial degeneration, and collapsed thermogenic capacity, by the virtue of mitochondrial dysfunction, devascularization, autophagy, and inflammation.
Collapse
Affiliation(s)
- Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mmabatho, South Africa
| | - Phiwayinkosi V. Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
- Department of Biochemistry and Microbiology, Faculty of Science and Agriculture, University of Zululand, KwaDlangezwa, South Africa
| | - Sinenhlanhla X. H. Mthembu
- Department of Biochemistry, North-West University, Mmabatho, South Africa
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Bongani B. Nkambule
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Sihle E. Mabhida
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Babalwa U. Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa
| | - Tawanda M. Nyambuya
- Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek, Namibia
| | | |
Collapse
|
46
|
Physiological and molecular mechanisms of cold-induced improvements in glucose homeostasis in humans beyond brown adipose tissue. Int J Obes (Lond) 2023; 47:338-347. [PMID: 36774412 DOI: 10.1038/s41366-023-01270-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/26/2023] [Accepted: 01/31/2023] [Indexed: 02/13/2023]
Abstract
Exposure to low ambient temperatures has previously been demonstrated to markedly improve glucose homeostasis in both rodents and humans. Although the brown adipose tissue is key in mediating these beneficial effects in rodents, its contribution appears more limited in humans. Hence, the exact tissues and underlying mechanisms that mediate cold-induced improvements in glucose homeostasis in humans remain to be fully established. In this review, we evaluated the response of the main organs involved in glucose metabolism (i.e. pancreas, liver, (white) adipose tissue, and skeletal muscle) to cold exposure and discuss their potential contribution to cold-induced improvements in glucose homeostasis in humans. We here show that cold exposure has widespread effects on metabolic organs involved in glucose regulation. Nevertheless, cold-induced improvements in glucose homeostasis appear primarily mediated via adaptations within the skeletal muscle and (presumably) white adipose tissue. Since the underlying mechanisms remain elusive, future studies should be aimed at pinpointing the exact physiological and molecular mechanisms involved in humans. Nonetheless, cold exposure holds great promise as a novel, additive lifestyle approach to improve glucose homeostasis in insulin resistant individuals. Parts of this graphical abstract were created using (modified) images from Servier Medical Art, licensed under the Creative Commons Attribution 3.0 Unported License. TG = thermogenesis, TAG = triacylglycerol, FFA = free fatty acid, SLN = sarcolipin, UCP3 = uncoupling protein 3, β2-AR = beta-2 adrenergic receptor, SNS = sympathetic nervous system.
Collapse
|
47
|
Geoghegan G, Simcox J. SON-light activation of glucose regulation. Cell 2023; 186:238-240. [PMID: 36669471 PMCID: PMC10246588 DOI: 10.1016/j.cell.2022.12.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/20/2023]
Abstract
Body temperature maintenance is an important regulator of glucose homeostasis. In this issue of Cell, Meng et al. discover a regulatory axis in which light activation of photoreceptive retinal ganglia stimulates the supraoptic nucleus (SON) to inhibit brown adipose tissue (BAT) thermogenesis and impair glucose homeostasis. This could explain the impact of constant light exposure on metabolism.
Collapse
Affiliation(s)
- Gisela Geoghegan
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Judith Simcox
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
48
|
Saito M, Okamatsu-Ogura Y. Thermogenic Brown Fat in Humans: Implications in Energy Homeostasis, Obesity and Metabolic Disorders. World J Mens Health 2023:41.e26. [PMID: 36792089 DOI: 10.5534/wjmh.220224] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/08/2022] [Indexed: 01/27/2023] Open
Abstract
In mammals including humans, there are two types of adipose tissue, white and brown adipose tissues (BATs). White adipose tissue is the primary site of energy storage, while BAT is a specialized tissue for non-shivering thermogenesis to dissipate energy as heat. Although BAT research has long been limited mostly in small rodents, the rediscovery of metabolically active BAT in adult humans has dramatically promoted the translational studies on BAT in health and diseases. It is now established that BAT, through its thermogenic and energy dissipating activities, plays a role in the regulation of body temperature, whole-body energy expenditure, and body fatness. Moreover, increasing evidence has demonstrated that BAT secretes various paracrine and endocrine factors, which influence other peripheral tissues and control systemic metabolic homeostasis, suggesting BAT as a metabolic regulator, other than for thermogenesis. In fact, clinical studies have revealed an association of BAT not only with metabolic disorders such as insulin resistance, diabetes, dyslipidemia, and fatty liver, but also with cardiovascular diseases including hypertension and atherosclerosis. Thus, BAT is an intriguing tissue combating obesity and related metabolic diseases. In this review, we summarize current knowledge on human BAT, focusing its patho-physiological roles in energy homeostasis, obesity and related metabolic disorders. The effects of aging and sex on BAT are also discussed.
Collapse
Affiliation(s)
- Masayuki Saito
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.
| | - Yuko Okamatsu-Ogura
- Laboratory of Biochemistry, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
49
|
Wang C, Zhang X, Liu M, Qin S, He C, Liu Y, Huai J, Zhang Q, Wei Y, Yang H. Irisin participates in the beneficial effects of exercise in preventing gestational diabetes mellitus in overweight and obese pregnant women and a mouse model. Front Nutr 2023; 9:1034443. [PMID: 36741990 PMCID: PMC9892194 DOI: 10.3389/fnut.2022.1034443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 12/20/2022] [Indexed: 01/20/2023] Open
Abstract
Objective We aimed to explore whether irisin participates in the beneficial effects of exercise in preventing gestational diabetes mellitus (GDM) in overweight and obese pregnant women. Study design Sixty overweight and obese pregnant women each in the exercise and control groups were randomly selected from our previous randomized controlled trial. Eighteen obese model mice were generated and divided into exercise and control groups in which body weight, abdominal circumference, anal temperature, glucose tolerance test, and insulin tolerance test were recorded. The plasma irisin level, the expression of PGC-1α/FNDC5 and brown (UCP1) and beige adipose (CD137, TMEM26, and TBX-1) marker genes were detected in muscle and adipose tissue. Results In the human study, women in the exercise group had a significantly higher irisin level and lower insulin resistance level than those in the control group. Enhanced expression of beige adipose tissue marker genes (CD137, TMEM26, and TBX-1) in omental adipose tissue and the CD137 gene in subcutaneous adipose tissue were found in the exercise group compared to the control group. In a mouse model, body weight and abdominal circumference were decreased, while glucose homeostasis and insulin sensitivity were significantly improved, and anal temperature was elevated after exercise intervention. A significantly higher level of irisin was revealed in the exercise group after undergoing exercise treatment. The expression of the beige adipose marker genes CD137 and TBX-1 was significantly higher in the exercise group than in the control group in posterior subcutaneous adipose tissue from the inguinal area and interscapular adipose tissue respectively. Conclusion Our observations show that regular exercise during pregnancy can increase irisin levels, promote white fat beiging/browning, improve glucose homeostasis and enhance body energy expenditure, which may be one of the mechanisms by which exercise prevents GDM.
Collapse
|
50
|
Takeda Y, Harada Y, Yoshikawa T, Dai P. Mitochondrial Energy Metabolism in the Regulation of Thermogenic Brown Fats and Human Metabolic Diseases. Int J Mol Sci 2023; 24:ijms24021352. [PMID: 36674862 PMCID: PMC9861294 DOI: 10.3390/ijms24021352] [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: 12/23/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Brown fats specialize in thermogenesis by increasing the utilization of circulating blood glucose and fatty acids. Emerging evidence suggests that brown adipose tissue (BAT) prevents the incidence of obesity-associated metabolic diseases and several types of cancers in humans. Mitochondrial energy metabolism in brown/beige adipocytes regulates both uncoupling protein 1 (UCP1)-dependent and -independent thermogenesis for cold adaptation and the utilization of excess nutrients and energy. Many studies on the quantification of human BAT indicate that mass and activity are inversely correlated with the body mass index (BMI) and visceral adiposity. Repression is caused by obesity-associated positive and negative factors that control adipocyte browning, de novo adipogenesis, mitochondrial energy metabolism, UCP1 expression and activity, and noradrenergic response. Systemic and local factors whose levels vary between lean and obese conditions include growth factors, inflammatory cytokines, neurotransmitters, and metal ions such as selenium and iron. Modulation of obesity-associated repression in human brown fats is a promising strategy to counteract obesity and related metabolic diseases through the activation of thermogenic capacity. In this review, we highlight recent advances in mitochondrial metabolism, thermogenic regulation of brown fats, and human metabolic diseases.
Collapse
Affiliation(s)
- Yukimasa Takeda
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| | - Yoshinori Harada
- Department of Pathology and Cell Regulation, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshikazu Yoshikawa
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Louis Pasteur Center for Medical Research, 103-5 Tanaka-Monzen-cho, Sakyo-ku, Kyoto 606-8225, Japan
| | - Ping Dai
- Department of Cellular Regenerative Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Correspondence: (Y.T.); (P.D.); Tel.: +81-75-251-5444 (Y.T.); +81-75-251-5135 (P.D.)
| |
Collapse
|