1
|
Wang H, Qin Y, Niu J, Chen H, Lu X, Wang R, Han J. Evolving perspectives on evaluating obesity: from traditional methods to cutting-edge techniques. Ann Med 2025; 57:2472856. [PMID: 40077889 PMCID: PMC11912248 DOI: 10.1080/07853890.2025.2472856] [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: 10/14/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Objective: This review examines the evolution of obesity evaluation methods, from traditional anthropometric indices to advanced imaging techniques, focusing on their clinical utility, limitations, and potential for personalized assessment of visceral adiposity and associated metabolic risks. Methods: A comprehensive analysis of existing literature was conducted, encompassing anthropometric indices (BMI, WC, WHR, WHtR, NC), lipid-related metrics (LAP, VAI, CVAI, mBMI), and imaging technologies (3D scanning, BIA, ultrasound, DXA, CT, MRI). The study highlights the biological roles of white, brown, and beige adipocytes, emphasizing visceral adipose tissue (VAT) as a critical mediator of metabolic diseases. Conclusion: Although BMI and other anthropometric measurements are still included in the guidelines, indicators that incorporate lipid metabolism information can more accurately reflect the relationship between metabolic diseases and visceral obesity. At the same time, the use of more modern medical equipment, such as ultrasound, X-rays, and CT scans, allows for a more intuitive assessment of the extent of visceral obesity.
Collapse
Affiliation(s)
- Heyue Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaxin Qin
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jinzhu Niu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Haowen Chen
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinda Lu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Rui Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jianli Han
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| |
Collapse
|
2
|
Yaligar J, Rengaraj A, Le GTT, Leow MKS, Eriksson JG, Ashokkumar B, Velan SS. Fatty Acylcarnitine Metabolism in Brown/Beige and White Fats by 13C HRMAS NMR Spectroscopy With Metabolic Interventions. NMR IN BIOMEDICINE 2025; 38:e70040. [PMID: 40230060 DOI: 10.1002/nbm.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/16/2025]
Abstract
White adipose tissue (WAT) and brown adipose tissue (BAT) have distinct structural and physiological characteristics and serve opposing functions in the body. WAT primarily stores energy, whereas BAT is metabolically active and positively influences metabolic health, contributing to energy expenditure, reduced fat accumulation and enhanced mitochondrial metabolism. Recently, both classical BAT and beige fat (or inducible/recruitable BAT) that arises from the browning of WAT have attracted clinical interest as potential targets for improving mitochondrial metabolism and managing obesity-related metabolic disorders. Currently, there is a lack of specific metabolic biomarkers for characterizing classical BAT and beige fat tissues, which are essential for evaluating mitochondrial oxidative metabolism and screening browning agents for therapeutic use. In this study, we investigated the potential metabolic biomarker fatty acylcarnitine in interscapular BAT (iBAT) from the interscapular region and beige adipose tissue from the inguinal region of the animal using ex vivo 13C high-resolution magic angle spinning (HRMAS) NMR spectroscopy. We examined how mitochondrial oxidative metabolism was altered in response to a high-fat diet (HFD) and how it was restored through iBAT activation using stimuli such as cold exposure and β3-adrenergic receptor (β3-AR) agonist, CL-316,243 treatment. We identified fatty acylcarnitine as a potential metabolic marker present in iBAT and beige tissues, whereas it was absent in iWAT. The concentration of fatty acylcarnitine significantly decreased in HFD-fed animals due to impaired lipolysis resulting in inefficient shuttling of fatty acids for mitochondrial β oxidation. However, its concentration increased or was restored in response to iBAT activation through either β3-AR agonist treatment or cold exposure, indicating a high-energy metabolic state with enhanced mitochondrial metabolism in iBAT. Fatty acylcarnitine shows promise as a biomarker for evaluating mitochondrial metabolism and for screening potential browning agents and nutraceuticals capable of inducing the browning of WAT.
Collapse
Affiliation(s)
- Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Anantharaj Rengaraj
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Department of Genetic Engineering, School of Biotechnology, Madurai Kamaraj University, Madurai, India
| | - Giang Thi Thu Le
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
| | - Melvin Khee-Shing Leow
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Cardiovascular and Metabolic Diseases Program, Duke-NUS Medical School, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- Division of Medicine, Department of Endocrinology, Tan Tock Seng Hospital, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | - S Sendhil Velan
- Institute for Human Development and Potential, Agency for Science Technology and Research (A*STAR), Singapore
- Laboratory of Molecular Imaging, Institute of Bioengineering and Bioimaging, A*STAR, Singapore
- Human Magnetic Resonance Centre, Institute for Applied Life Sciences, University of Massachusetts Amherst, Amherst, Massachusetts, USA
| |
Collapse
|
3
|
Han X, Zeng X, Gao S, Zhang Q, Zheng K, Yang H, Hu B, Ding C. Adipose-targeted nanohybrid as a browning inducer for synergistic hyperthermia-pharmacotherapy of obesity. J Colloid Interface Sci 2025; 687:540-551. [PMID: 39978259 DOI: 10.1016/j.jcis.2025.02.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Inducing adipose browning to increase energy expenditure has recently emerged as a promising approach for antiobesity treatment. However, its therapeutic efficacy is often limited by poor adipose-targeted drug delivery and suboptimal browning efficiency. To address these challenges, an adipose-targeting aptamer (Apt8) and browning agent resveratrol (Res) were used to construct an Apt-modified and Res-loaded degradable mesoporous silica-coated Au nanorods nanocarriers (NC), termed Res@NC@Apt8, achieving adipose-targeted hyperthermia-pharmacotherapy. Upon internalization by adipocytes, laser irradiation induces mild local hyperthermia (LHT) via Res@NC@Apt8, triggering calcium ion (Ca2+) influx. Simultaneously, the interaction of the nanohybrid with local glutathione (GSH) releases Res. The dual mechanisms activate the adenosine 5'-monophosphate-activated protein kinase (AMPK) pathway, reduce the lipid droplet content, enhance mitochondrial biogenesis, and accelerate metabolism, thereby synergistically promoting adipose browning. Intravenous Res@NC@Apt8 administration in obese mice significantly drives adipose reduction and further achieves excellent antiobesity therapeutic efficacy. This synergistic treatment achieves a superior weight reduction of 17.2% compared with 6.9% and 10.6% achieved using LHT and pharmacotherapy alone, respectively. This study introduces a novel strategy for achieving activatable LHT and drug release for synergetic obesity treatment.
Collapse
Affiliation(s)
- Xiaoyang Han
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Xiaohan Zeng
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Shiwen Gao
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Qian Zhang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Ke Zheng
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Huiwen Yang
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Bo Hu
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| | - Caifeng Ding
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, Ministry of Education, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao 266042, PR China.
| |
Collapse
|
4
|
Simoes MR, Bombassaro B, Gallo-Ferraz AL, Nogueira PAS, Monfort-Pires M, Zanesco AM, Valdivieso-Rivera F, Nogueira GAS, Sponton CH, Castilho RF, Velloso LA. Balb/c mice are protected from glucose and acute cold intolerance. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167725. [PMID: 40023454 DOI: 10.1016/j.bbadis.2025.167725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/21/2025] [Accepted: 02/12/2025] [Indexed: 03/04/2025]
Abstract
The brown adipose tissue is a potential target for interventions aimed at treating obesity and other metabolic disorders. Both genetic and environmental factors are known to regulate brown adipose tissue function and exploring the interaction between these factors could unveil new mechanisms involved in the regulation of thermogenesis. In this study, we evaluated three genetically distinct mice strains submitted to two environmental factors known to modulate brown adipose tissue function, namely, cold exposure and the consumption of a high-fat diet. The comparison of Balb/c, C57BL/6, and Swiss mice revealed that Balb/c mice were the most glucose-tolerant and the most cold-tolerant. In addition, Balb/c presented the greatest brown adipose tissue oxygen consumption, which was independent of differences in uncoupling protein 1 expression and function. The search for uncoupling protein 1-independent mechanisms that could explain the greatest cold tolerance of Balb/c mice resulted in the identification of the N-acyl amino acid regulator, PM20D1, which had a greater gene expression in the brown adipose tissue of Balb/c mice as compared to the other two strains. The immunoneutralization of PM20D1 in Balb/c mice, resulted in increased blood glucose levels and worsening of cold tolerance. In addition, the in silico knockout of Pm20d1 impacted several metabolic processes, including thermogenesis, glucose tolerance, and insulin sensitivity. In conclusion, Balb/c mice are protected from glucose and acute cold intolerance, independently of the diet. We propose that PM20D1, in an uncoupling protein 1-independent fashion, can have an important role in this protection.
Collapse
Affiliation(s)
- Marcela R Simoes
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Ana Luisa Gallo-Ferraz
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Pedro A S Nogueira
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | | | - Ariane M Zanesco
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Fernando Valdivieso-Rivera
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas (UNICAMP), Campinas, São Paulo 13083-862, Brazil
| | - Guilherme A S Nogueira
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil
| | - Carlos H Sponton
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; Department of Structural and Functional Biology, Institute of Biology (IB), University of Campinas (UNICAMP), Campinas, São Paulo 13083-862, Brazil
| | - Roger F Castilho
- Department of Pathology, School of Medical Sciences, University of Campinas (UNICAMP), Campinas, SP 13083-888, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo 13083-864, Brazil; National Institute of Science and Technology on Neuroimmunomodulation, Campinas, São Paulo 13083-864, Brazil.
| |
Collapse
|
5
|
Wei G, Shen FJ, Liu JL, Zhao JH, Yang FY, Feng RQ, Lu J, Zhang CY, Wang FW, Chen BD, Ding X, Yang JK. Uncoupling protein 1 deficiency leads to transcriptomic differences in livers of pregnancy female mice and aggravates hepatic steatosis. Arch Biochem Biophys 2025; 768:110395. [PMID: 40122441 DOI: 10.1016/j.abb.2025.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Pregnancy requires the coordination of metabolically active organs to support maternal nutrition and fetal growth. However, the metabolic cross-talk between adipose tissue and liver in females during pregnancy is still less clear. In this study, we evaluated the metabolic adaptations and phenotypes of liver in response to pregnancy-associated metabolic stress, particularly in the context of genetic ablation of Uncoupling protein 1 (Ucp1)-mediated catabolic circuit. Our results revealed that Ucp1 deficiency (UCP1 knockout, KO) mice during late pregnancy exhibited significantly deteriorated metabolic phenotypes, including hepatic steatosis and whole-body glucose and lipid homeostasis, as compared to Ucp1 deficiency or normal pregnancy mice. However, non-pregnant Ucp1 deficiency mice displayed nearly normal metabolic phenotypes and structure alterations similar to those of littermate controls. Moreover, transcriptomic analyses by RNA sequencing (RNA-seq) clearly revealed that Ucp1 deficiency led to a significant liver metabolic remodeling of differentially express genes (DEGs) before and especially during pregnancy. Consistently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses demonstrated the potential altered functions and signaling pathways, including metabolic dysfunctions in ribosome, oxidative phosphorylation, etc. Importantly, as derived from trend analyses of DEGs, our results further revealed the distinct expression pattern of each subcluster, which coincided with potential biological functions and relevant signaling pathways. The findings in the present study might provide valuable insights into the molecular mechanism of metabolic dysfunction-associated fatty liver disease (MAFLD) during pregnancy. Additionally, our data may provide a novel animal model of MAFLD, thus facilitating its potential therapies. NEW & NOTEWORTHY: Genetic ablation of Ucp1 during pregnancy increases hepatic steatosis and deteriorated whole-body glucose and lipid homeostasis. Moreover, changes in hepatic gene expression are closely associated with metabolic dysfunctions in ribosome and oxidative phosphorylation. This work highlights the therapeutic potential of targeting UCP1- mediated catabolic circuit between adipose and liver during pregnancy, and the utility of RNA-seq analysis to reveal valuable information for the distinct expression pattern of each subcluster that contribute to pregnancy-dependent MASLD progression.
Collapse
Affiliation(s)
- Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| | - Feng-Jie Shen
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jun-Li Liu
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Jian-Hua Zhao
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Fang-Yuan Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Ruo-Qi Feng
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Chen-Yang Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Feng-Wei Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Bei-Dong Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100005, China.
| | - Xin Ding
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100020, China.
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
6
|
Bat-Erdene B, He M, Dong J, Li Y, Ta D. Therapeutic Effects of Different Ultrasound Intensity Stimulation on Brown Adipose Tissue for the Treatment of Type 2 Diabetes. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:830-840. [PMID: 39924417 DOI: 10.1016/j.ultrasmedbio.2025.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 02/11/2025]
Abstract
Type 2 diabetes (T2D) is a persistent illness that has a high incidence rate. Still, there is no conclusive evidence on effectively improving blood sugar levels in patients through physical therapy. This study examined the regulatory effects of different intensities of low-intensity pulsed ultrasound (LIPUS) on T2D by stimulating brown adipose tissue (BAT). Eight-week-old C57BL/6J mice were divided into six groups (n = 10 per group): Control sham (C-Sham), Control-LIPUS (C-LIPUS), T2D-sham (T2D-Sham), T2D groups treated with LIPUS at spatial average-temporal-average intensity (Isata) of 60mW/cm² (T2D-L-60), 80mW/cm² (T2D-L-80), and 100mW/cm² (T2D-L-100). T2D models were induced by intraperitoneal injection of 40 mg/kg streptozotocin (STZ) three times after 12 wks of high-fat diet (HFD). The T2D-LIPUS group received LIPUS stimulation for 20 minutes per day for 6 weeks. The LIPUS stimulation had a duty cycle of 20%, a frequency of 1 MHz, and Isata of 60mW/cm², 80mW/cm², 100mW/cm². Subsequently, glucose tolerance tests (GTT) and insulin tolerance tests (ITT) were performed, and body fat content in mice was analyzed using nuclear magnetic resonance (NMR). Metabolic changes were monitored using metabolic cages. The results indicated that 80mW/cm² intensity level significantly improved glucose tolerance, insulin sensitivity, and metabolic function after LIPUS exposure. Significant reductions in body fat content and enhanced thermogenesis were observed, highlighting the potential of LIPUS in T2D management. This provides the basis for the dose study of LIPUS in the treatment of T2D.
Collapse
Affiliation(s)
- Badamgarav Bat-Erdene
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Min He
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China.
| | - Jingsong Dong
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Ying Li
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China
| | - Dean Ta
- Department of Biomedical Engineering, School of Information Science and Technology, Fudan University, Shanghai, China; Academy for Engineering and Technology, Fudan University, Shanghai, China; State Key Laboratory of Integrated Chips and Systems, Fudan University, Shanghai, China; Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Yoneshiro T, Matsushita M, Sakai J, Saito M. Brown fat thermogenesis and cold adaptation in humans. J Physiol Anthropol 2025; 44:11. [PMID: 40259336 DOI: 10.1186/s40101-025-00391-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/30/2025] [Indexed: 04/23/2025] Open
Abstract
Brown adipose tissue (BAT) is a site of non-shivering thermogenesis (NST) in mammals. Since the rediscovery of BAT in adult humans, there has been a remarkable advance in human BAT researches, revealing the significant roles of this thermogenic tissue in cold-induced NST and cold adaptation. Cold stress influences BAT in various time spans: acute cold exposure promptly activates BAT to induce NST, which contributes to immediate maintenance of body temperature. Prolonged cold exposure recruits BAT, resulting in increased capacity of NST and improved cold tolerance. Such BAT adaptation not only occurs in the exposed individual but also is passed on to the next generation, probably via the paternal lineage. As such, BAT plays a role in acute, chronic, and transgenerational adaptation to cold environment in humans.
Collapse
Affiliation(s)
- Takeshi Yoneshiro
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 980 - 8575, Japan.
| | - Mami Matsushita
- Department of Nutrition, School of Nursing and Nutrition, Tenshi College, Sapporo, Hokkaido, 065 - 0013, Japan
| | - Juro Sakai
- Division of Molecular Physiology and Metabolism, Tohoku University Graduate School of Medicine, Sendai, Miyagi, 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
| |
Collapse
|
8
|
Jiao W, Park WY, Kim YI, Kim JH, Kim B, Song G, Park JY, Jung SJ, Kwak HJ, Choe SK, Lee JH, Um JY. Browning of inguinal white adipose tissue by the novel lignan (-)-secoisolariciresinol 4-O-methyl ether attenuates diet-induced obesity through mitochondrial and peroxisomal activation. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119952. [PMID: 40194601 DOI: 10.1016/j.bbamcr.2025.119952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/09/2025]
Abstract
Studies indicate that the induction and activation of brown and beige adipocytes, which can enhance energy expenditure, may be beneficial for managing obesity and its associated diseases. This study investigated whether a novel lignan (-)-secoisolariciresinol 4-O-methyl ether (S4M) obtained from arctigenin inhibited diet-induced obesity by the browning of white adipose tissue (WAT). S4M treatment inhibited adipogenesis and lipid accumulation in white-induced 3T3-L1 adipocytes and in zebrafish embryonic development. Moreover, S4M treatment promoted browning in white adipocytes by increasing TOM20, UCP1, and PGC1α protein levels and consequently upregulating the mitochondrial content. S4M treatment significantly promoted mitochondrial fission by increasing the expression of DRP1. Furthermore, it enhanced peroxisomal biogenesis and function by inducing PEX13, ACOX1, and catalase. Mdivi-1, a mitochondrial dynamics inhibitor, diminished the browning effect of white adipocytes by the S4M treatment. This study found that S4M treatment inhibited weight gain in high-fat diet-induced obese mice, decreased the weight of WAT, and increased the abundance and function of mitochondria and peroxisomes in inguinal WAT, suggesting that S4M treatment could increase energy expenditure. The results suggest that S4M has potential as a therapeutic agent for combating obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Wenjun Jiao
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woo Yong Park
- Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yong-Il Kim
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Jin-Hyung Kim
- Department of Biomedical and Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Beomsu Kim
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Gahee Song
- Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ja Yeon Park
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Se Jin Jung
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyun Jeong Kwak
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
| | - Seong-Kyu Choe
- Department of Microbiology, Wonkwang University School of Medicine, Iksan 54538, Republic of Korea
| | - Jong-Hyun Lee
- College of Pharmacy, Dongduk Women's University, 60 Hwarang-ro 13-gil, Seongbuk-gu, Seoul, 02748, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; Kyung Hee Institute of Convergence Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Department of Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
9
|
Xu Y, Chen Y, Bai N, Su Y, Ye Y, Zhang R, Yang Y, Liu C, Hu C, Pan J. Deubiquitinating enzyme USP2 regulates brown adipose tissue thermogenesis via controlling EBF2 stabilization. Mol Metab 2025; 96:102139. [PMID: 40189098 DOI: 10.1016/j.molmet.2025.102139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/23/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
OBJECTIVE The activation of brown adipose tissue (BAT) promotes energy expenditure is recognized as a promising therapeutic strategy for combating obesity. The deubiquitinating enzyme family members are widely involved in the process of energy metabolism. However, the specific deubiquitinating enzyme member that affects the BAT thermogenesis remains largely unexplored. METHODS Adeno-associated virus, lentivirus and small molecule inhibitor were applied to generate USP2 gain- or loss-of-function both in vivo and in vitro. OxyMax comprehensive laboratory animal monitoring system, seahorse and transmission electron microscopy were used to determine the energy metabolism. Quantitative proteomics, immunofluorescence staining and co-immunoprecipitation were performed to reveal the potential substrates of USP2. RESULTS USP2 is upregulated upon thermogenic activation in adipose, and has a close correlation with UCP1 mRNA levels in human adipose tissue. BAT-specific Usp2 knockdown or systemic USP2 inhibition resulted in impaired thermogenic programs both in vivo and in vitro. Conversely, overexpression of Usp2 in BAT conferred protection against high-fat diet-induced obesity and associated metabolic disorders. Proteome-wide analysis identified EBF2 as the substrate of USP2 that mediates the thermogenic function of USP2 in BAT. CONCLUSIONS Our data demonstrated the vital role of USP2 in regulating BAT activation and systemic energy homeostasis. Activation of USP2-EBF2 interaction could be a potential therapeutic strategy against obesity.
Collapse
Affiliation(s)
- Yuejie Xu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ying Chen
- Jinzhou Medical University Graduate Training Base (Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine), Jinzhou, 121001, China
| | - Ningning Bai
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yingying Su
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yafen Ye
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Rong Zhang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Caizhi Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Cheng Hu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Jiemin Pan
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Key Clinical Center for Metabolic Disease, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| |
Collapse
|
10
|
Zhou YQ, Chang XY, Yang L, Pan D, Huang HY. Loss of lysyl oxidase in adipose tissue ameliorates metabolic inflexibility induced by high-fat diet. Obesity (Silver Spring) 2025; 33:720-731. [PMID: 40025831 DOI: 10.1002/oby.24253] [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: 09/12/2024] [Revised: 11/23/2024] [Accepted: 12/31/2024] [Indexed: 03/04/2025]
Abstract
OBJECTIVE Systemic administration of β-aminopropionitrile to inhibit lysyl oxidase (Lox) activity improves metabolism, but it exhibits a broad spectrum of effects. Clarification of the role of Lox in adipose tissue metabolism under high-fat diet (HFD) conditions is needed. METHODS Mice with adipose tissue knockout of Lox (LoxAKO) and wild-type mice were subjected to a 16-week HFD regimen. A detailed evaluation encompassing adipose tissue, hepatic function, and systemic metabolism was conducted. RNA sequencing analysis was used to unravel the intricate mechanisms behind the metabolic enhancements in LoxAKO mice. RESULTS Compared with the control, although there was no difference in body weight, LoxAKO mice exhibited an improved metabolic phenotype, including enhanced insulin sensitivity, improved glucose tolerance, and reduced liver steatosis, along with reduced adipose tissue inflammation and fibrosis. LoxAKO mice showed increased thermogenic activity in brown adipose tissue with increased uncoupling protein 1 (UCP1) expression and oxygen consumption rate. Additionally, RNA sequencing analysis revealed that adipose deletion of Lox might facilitate the metabolic processing of glucose, branched-chain amino acids, and fatty acids in brown adipose tissue. CONCLUSIONS These findings indicate that adipocyte Lox deletion improves metabolic adaptability under an HFD, highlighting Lox as a promising therapeutic target for obesity-associated metabolic disorders.
Collapse
Affiliation(s)
- Yun-Qian Zhou
- 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, People's Republic of China
| | - Xin-Yue Chang
- 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, People's Republic of China
| | - Lei Yang
- 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, People's Republic of 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, People's Republic of China
| | - Hai-Yan 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, People's Republic of China
| |
Collapse
|
11
|
Sugino T, Taguchi K, Yanase T, Unno R, Ando R, Yasui T. Brown adipose tissue detection using positron emission tomography could help reduce urolithiasis risk. Urolithiasis 2025; 53:64. [PMID: 40167829 DOI: 10.1007/s00240-025-01739-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025]
Abstract
Urolithiasis is associated with metabolic syndrome, and reactivation of brown adipose tissue (BAT) may improve metabolic syndrome. In this study, we aimed to evaluate the association of BAT, as detected using positron emission tomography-computed tomography (PET-CT), with urolithiasis in humans. This single center retrospective cohort study involved patients who underwent PET-CT for cancer screening as part of a medical checkup between January 2006 and December 2020. We obtained data on participant demographics, presence of urolithiasis, and metabolic factors (such as obesity, hypertension, diabetes, and hyperlipidemia) from 182 medical records. BAT data and calcification of the abdominal aorta (CAA) rate, determined using abdominal CT, were also recorded. Any association between urolithiasis and other factors was evaluated using logistic regression analysis. Body mass index was higher in participants with BAT than in those without BAT (26.92 vs. 22.86 kg/m2, p = 0.001). Participants with BAT had less urolithiasis and a lower CAA rate than those without BAT (10% vs. 37%, p = 0.031 and 50% vs. 79.6%, p = 0.008, respectively). Metabolic factors between the groups were similar (45.0% vs. 48.8%, p = 0.936). Furthermore, BAT and age were associated with a decreased odds ratio (OR) for urolithiasis (OR = 0.186, p = 0.037 and OR = 0.959, p = 0.02, respectively). We demonstrated that high BAT content is associated with a low risk of urolithiasis and CAA. Our findings may contribute to the development of novel preventive methods for urolithiasis.
Collapse
Affiliation(s)
- Teruaki Sugino
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
- Department of Urology, Nagoya City University East Medical Center, Nagoya, Japan
| | - Kazumi Taguchi
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Takahiro Yanase
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Rei Unno
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryosuke Ando
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
12
|
Jones SA, Sowton AP, Lacabanne D, King MS, Palmer SM, Zögg T, Pardon E, Steyaert J, Ruprecht JJ, Kunji ERS. Proton conductance by human uncoupling protein 1 is inhibited by purine and pyrimidine nucleotides. EMBO J 2025; 44:2353-2365. [PMID: 40021843 PMCID: PMC12000319 DOI: 10.1038/s44318-025-00395-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/03/2025] Open
Abstract
Uncoupling protein 1 (UCP1, SLC25A7) is responsible for the thermogenic properties of brown adipose tissue. Upon fatty acid activation, UCP1 facilitates proton leakage, dissipating the mitochondrial proton motive force to release energy as heat. Purine nucleotides are considered to be the only inhibitors of UCP1 activity, binding to its central cavity to lock UCP1 in a proton-impermeable conformation. Here we show that pyrimidine nucleotides can also bind and inhibit its proton-conducting activity. All nucleotides bound in a pH-dependent manner, with the highest binding affinity observed for ATP, followed by dTTP, UTP, GTP and CTP. We also determined the structural basis of UTP binding to UCP1, showing that binding of purine and pyrimidine nucleotides follows the same molecular principles. We find that the closely related mitochondrial dicarboxylate carrier (SLC25A10) and oxoglutarate carrier (SLC25A11) have many cavity residues in common, but do not bind nucleotides. Thus, while UCP1 has evolved from dicarboxylate carriers, no selection for nucleobase specificity has occurred, highlighting the importance of the pH-dependent nucleotide binding mechanism mediated via the phosphate moieties.
Collapse
Affiliation(s)
- Scott A Jones
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Alice P Sowton
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Denis Lacabanne
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Martin S King
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Shane M Palmer
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Thomas Zögg
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050, Brussels, Belgium
| | - Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050, Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, B-1050, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, B-1050, Brussels, Belgium
| | - Jonathan J Ruprecht
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK
| | - Edmund R S Kunji
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Keith Peters Building, Cambridge, CB2 0XY, UK.
| |
Collapse
|
13
|
Tang X, Zhang B, Xie P, Wei Y, Qiu Y, Yi X, Zhang Z, She M, Sun X, Wang S. Dexamethasone-induced whitening of rabbit brown adipose tissue: leptin resistance and mitochondrial dysfunction. BMC Genomics 2025; 26:326. [PMID: 40165063 PMCID: PMC11959718 DOI: 10.1186/s12864-025-11502-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Research on the effects of exogenous glucocorticoids on brown adipose tissue (BAT) is crucial for understanding how these hormones can induce metabolic disorders such as obesity. In this study, we explored the effects of glucocorticoids, specifically dexamethasone (Dex), on the metabolic transformation of BAT to white adipose tissue (WAT). RESULTS Our results indicate a significant whitening shift in BAT upon Dex treatment, characterized by increased lipid deposition, decreased mitochondrial density, a significant decline in cellular ATP content, and reduced expression of mitochondrial markers. We demonstrate the crucial role of leptin resistance in mediating mitochondrial function through the overexpression and inhibition of LEPR. CONCLUSIONS Our results suggest the role of leptin resistance in regulating of mitochondrial biogenesis and energy metabolism in glucocorticoid-induced brown adipose whiteness.
Collapse
Affiliation(s)
- Xiaoqin Tang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Beibei Zhang
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Puhang Xie
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yanpei Wei
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yanbo Qiu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Xiaohua Yi
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Ziru Zhang
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Muzi She
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Xiuzhu Sun
- College of Grassland Agriculture, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China.
| | - Shuhui Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China.
| |
Collapse
|
14
|
Duerre DJ, Hansen JK, John SV, Jen A, Carrillo ND, Bui H, Bao Y, Fabregat M, Catrow JL, Chen LY, Overmyer KA, Shishkova E, Pearce Q, Keller MP, Anderson RA, Cryns VL, Attie AD, Cox JE, Coon JJ, Fan J, Galmozzi A. Haem biosynthesis regulates BCAA catabolism and thermogenesis in brown adipose tissue. Nat Metab 2025:10.1038/s42255-025-01253-6. [PMID: 40133548 DOI: 10.1038/s42255-025-01253-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
The distinctive colour of brown adipose tissue (BAT) is attributed to its high content of haem-rich mitochondria. However, the mechanisms by which BAT regulates intracellular haem levels remain largely unexplored. Here we demonstrate that haem biosynthesis is the primary source of haem in brown adipocytes. Inhibiting haem biosynthesis results in an accumulation of the branched-chain amino acids (BCAAs) valine and isoleucine, owing to a haem-associated metabolon that channels BCAA-derived carbons into haem biosynthesis. Haem synthesis-deficient brown adipocytes display reduced mitochondrial respiration and lower UCP1 levels than wild-type cells. Although exogenous haem supplementation can restore intracellular haem levels and mitochondrial function, UCP1 downregulation persists. This sustained UCP1 suppression is linked to epigenetic regulation induced by the accumulation of propionyl-CoA, a byproduct of disrupted haem synthesis. Finally, disruption of haem biosynthesis in BAT impairs thermogenic response and, in female but not male mice, hinders the cold-induced clearance of circulating BCAAs in a sex-hormone-dependent manner. These findings establish adipose haem biosynthesis as a key regulator of thermogenesis and sex-dependent BCAA homeostasis.
Collapse
Affiliation(s)
- Dylan J Duerre
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julia K Hansen
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Steven V John
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
- Morgridge Institute for Research, Madison, WI, USA
| | - Annie Jen
- Integrated Program in Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Noah D Carrillo
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Molecular and Environmental Toxicology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Hoang Bui
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Nutrition and Metabolism Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Yutong Bao
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Matias Fabregat
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - J Leon Catrow
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Li-Yu Chen
- Graduate Program in Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Katherine A Overmyer
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Evgenia Shishkova
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Quentinn Pearce
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Richard A Anderson
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Vincent L Cryns
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - James E Cox
- Metabolomics Core Research Facility, University of Utah, Salt Lake City, UT, USA
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Joshua J Coon
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- National Center for Quantitative Biology of Complex Systems, Madison, WI, USA
| | - Jing Fan
- Morgridge Institute for Research, Madison, WI, USA
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA
- Department of Medical Microbiology & Immunology, University of Wisconsin-Madison, Madison, WI, USA
| | - Andrea Galmozzi
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
15
|
Antonopoulou S. Platelet-Activating Factor-Induced Inflammation in Obesity: A Two-Sided Coin of Protection and Risk. Cells 2025; 14:471. [PMID: 40214425 PMCID: PMC11987740 DOI: 10.3390/cells14070471] [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: 03/06/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity, marked by excessive fat accumulation, especially abdominal, is a global health concern with significant public impact. While obesity-associated chronic unresolved inflammation contributes to metabolic dysfunctions, acute inflammation supports healthy adipose tissue remodeling and expansion. Platelet-activating factor (PAF), a "primitive" signaling molecule, is among the key mediators involved in the acute phase of inflammation and in various pathophysiological processes. This article explores the role of PAF in fat accumulation and obesity by reviewing experimental data from cell cultures, animals, and humans. It proposes an emerging biochemical mechanism in an attempt to explain its dual role in the healthy and obese adipose tissue, including also data on PAF's potential involvement in epigenetic mechanisms that may be linked to the "obesity memory". Finally, it highlights the potential of natural PAF modulators in promoting functional adipose tissue, thermogenesis, and obesity prevention through a healthy lifestyle, including a Mediterranean diet rich in PAF weak agonists/PAF receptor antagonists and regular exercise, which help maintain controlled PAF levels. Conversely, in cases of obesity-related systemic inflammation with excessive PAF levels, potent PAF inhibitors like ginkgolide B and rupatadine may help mitigate metabolic dysfunctions with PAFR antagonists potentially enhancing their effects synergistically.
Collapse
Affiliation(s)
- Smaragdi Antonopoulou
- Department of Nutrition and Dietetics, School of Health Sciences and Education, Harokopio University, 17671 Athens, Greece
| |
Collapse
|
16
|
Miao Q, Wang Y, Zhang Q, Wu W, Yu Y, Zeng F, Yang Y, Zuo C, Guan Y, Liew CW, Blüher M, Li Y, Wang X. Serum secreted EMC10 (scEMC10) levels are inversely associated with metabolically active brown adipose tissue in humans. Int J Obes (Lond) 2025:10.1038/s41366-025-01744-2. [PMID: 40102590 DOI: 10.1038/s41366-025-01744-2] [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: 05/02/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND/OBJECTIVES Secreted endoplasmic reticulum membrane complex subunit 10 (scEMC10) has been implicated in obesity in mice and humans. In this study, the associations of serum scEMC10 levels with thermoneutrality-modulated brown adipose tissue (BAT) activity and thyroid hormone (TH)-dependent thermogenesis were investigated in humans. SUBJECTS/METHODS Serum scEMC10 levels were measured in participants from multiple cohorts using enzyme-linked immunosorbent assay, including participants with or without active BAT determined by PET-CT scanning, participants with positive BAT before and after thermoneutrality, and patients with hyperthyroidism before and after anti-thyroid drug (ATD) treatment. The difference in serum scEMC10 between participants with positive or negative BAT, and the changes of serum scEMC10 in participants with positive BAT before and after thermoneutrality and in patients with Grave's disease-caused hyperthyroidism before and after ATD treatment were determined. RESULTS PET-CT scan with 18F-FDG indicated participants with positive BAT were significantly younger and leaner than ones with negative BAT. There was, however, no significant difference in serum scEMC10 between the two groups. Serum scEMC10 levels in participants with positive BAT were significantly elevated by 2-h thermoneutrality (p = 0.0017), concomitant with disappearance of active BAT. No significant association of serum scEMC10 with serum levels of either TSH, FT3, or FT4 was observed in participants from both Chinese and White cohorts. ATD treatment normalized thyroid function and reduced the uptake of 18F-FDG into skeletal muscle of patients with hyperthyroidism. Serum scEMC10 concentration, however, remained unchanged in these patients before and after ATD treatment. CONCLUSIONS Serum scEMC10 levels are inversely associated with BAT activity in humans.
Collapse
Affiliation(s)
- Qing Miao
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yahao Wang
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiongyue Zhang
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wu
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yifei Yu
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fangfang Zeng
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yehong Yang
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chuantao Zuo
- PET Center & Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yihui Guan
- PET Center & Department of Nuclear Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chong Wee Liew
- Department of Physiology & Biophysics, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
- Medical Department III-Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig, Germany
| | - Yiming Li
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xuanchun Wang
- Department of Endocrinology, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
McLeod K, Datta V, Fuller S. Adipokines as Cardioprotective Factors: BAT Steps Up to the Plate. Biomedicines 2025; 13:710. [PMID: 40149686 PMCID: PMC11940801 DOI: 10.3390/biomedicines13030710] [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: 01/10/2025] [Revised: 03/07/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Cardiovascular disease is the leading cause of death throughout most of the industrialized world. Metabolic syndrome (MetS) and its associated pathologies are underlying factors in the etiology of cardiovascular disease, as well as a plethora of other maladies which cause excess morbidity and mortality. Adipose tissue (AT) has come to be regarded as a bona fide endocrine organ which secretes specific molecular entities constituting part of a complex web of inter-organ crosstalk that functions as a key determinant of whole-body metabolic phenotype. Brown adipose tissue (BAT) has classically been regarded as a thermogenic tissue exerting its metabolic effects primarily through its capacity to oxidize substrates decoupled from ATP resynthesis, thereby resulting in increased energy expenditure (EE) and heat production. However, in recent years, BAT has begun to receive attention as a secretory organ in its own right. The molecules secreted specifically by BAT have been termed "batokines", and currently available evidence supports the notion that batokines exert favorable metabolic effects on multiple organ systems. While maintenance of healthy body composition by conferring resistance to excessive adiposity is a rather obvious mechanism by which BAT operates via increased EE, effects on critical organs such as the heart remain unclear. This narrative review focuses on four types of batokines (FGF21, neuregulin 4, 12,13-diHOME, and BAT-derived microRNAs) for which evidence of modulation of cardiovascular function exists in the context of pathological states such as hypertension, atherosclerosis, and ischemia/reperfusion injury. Given the overwhelming burden of cardiometabolic disease, further study of the functions of BAT and its secretome is warranted and will intensify in the future.
Collapse
Affiliation(s)
- Keely McLeod
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| | - Victoria Datta
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
| | - Scott Fuller
- School of Kinesiology, University of Louisiana at Lafayette, Lafayette, LA 70506, USA; (K.M.); (V.D.)
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA
| |
Collapse
|
18
|
Dumesic PA, Wilensky SE, Bose S, Van Vranken JG, Gygi SP, Spiegelman BM. RBM43 controls PGC1α translation and a PGC1α-STING signaling axis. Cell Metab 2025; 37:742-757.e8. [PMID: 39965564 PMCID: PMC11885043 DOI: 10.1016/j.cmet.2025.01.013] [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: 01/30/2024] [Revised: 10/17/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Obesity is associated with systemic inflammation that impairs mitochondrial function. This disruption curtails oxidative metabolism, limiting adipocyte lipid metabolism and thermogenesis, a metabolically beneficial program that dissipates chemical energy as heat. Here, we show that PGC1α, a key governor of mitochondrial biogenesis, is negatively regulated at the level of its mRNA translation by the RNA-binding protein RBM43. RBM43 is induced by inflammatory cytokines and suppresses mitochondrial biogenesis in a PGC1α-dependent manner. In mice, adipocyte-selective Rbm43 disruption elevates PGC1α translation and oxidative metabolism. In obesity, Rbm43 loss improves glucose tolerance, reduces adipose inflammation, and suppresses activation of the innate immune sensor cGAS-STING in adipocytes. We further identify a role for PGC1α in safeguarding against cytoplasmic accumulation of mitochondrial DNA, a cGAS ligand. The action of RBM43 defines a translational regulatory axis by which inflammatory signals dictate cellular energy metabolism and contribute to metabolic disease pathogenesis.
Collapse
Affiliation(s)
- Phillip A Dumesic
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sarah E Wilensky
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Symanthika Bose
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Bruce M Spiegelman
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Sakaguchi K, Sugawara K, Hosokawa Y, Ito J, Morita Y, Mizuma H, Watanabe Y, Kimura Y, Aburaya S, Takahashi M, Izumi Y, Bamba T, Komada H, Yamada T, Hirota Y, Yoshida M, Nogami M, Murakami T, Ogawa W. Metformin-regulated glucose flux from the circulation to the intestinal lumen. COMMUNICATIONS MEDICINE 2025; 5:44. [PMID: 40033038 DOI: 10.1038/s43856-025-00755-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Through a retrospective analysis of existing FDG PET-MRI images, we recently demonstrated that metformin increases the accumulation of FDG in the intestinal lumen, suggesting that metformin stimulates glucose excretion into the intestine. However, the details of this phenomenon remain unclear. We here investigate the detailed dynamics of intestinal glucose excretion, including the rate of excretion and the metabolism of excreted glucose, in both the presence and absence of metformin. METHODS We quantified intestinal glucose excretion using newly developed FDG PET-MRI-based bioimaging in individuals with type 2 diabetes, both treated and untreated with metformin. The metabolism of excreted glucose was analyzed through mass spectrometry of fecal samples from mice intravenously injected with 13C-labeled glucose. RESULTS Continuous FDG PET/MRI image taking reveals that FDG is initially observed in the jejunum, suggesting its involvement in FDG excretion. Metformin-treated individuals excrete a significant amount of glucose (~1.65 g h-1 per body) into the intestinal lumen. In individuals not receiving metformin, a certain amount of glucose (~0.41 g h-1per body) is also excreted into the intestinal lumen, indicating its physiological importance. Intravenous injection of 13C-labeled glucose in mice increases the content of 13C in short-chain fatty acids (SCFAs) extracted from feces, and metformin increased the incorporation of 13C into SCFAs. CONCLUSIONS A previously unrecognized, substantial flux of glucose from the circulation to the intestinal lumen exists, which likely contributes to the symbiosis between gut microbiota and the host. This flux represents a potential target of metformin's action in humans.
Collapse
Affiliation(s)
- Kazuhiko Sakaguchi
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Community Medicine and Medical Education, Department of Social/Community Medicine and Health Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenji Sugawara
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yusei Hosokawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Jun Ito
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuko Morita
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Mizuma
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yasuyoshi Watanabe
- Laboratory for Pathophysiological and Health Science, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Yuichi Kimura
- Faculty of Informatics, Cyber Informatics Research Institute, Kindai University, Osaka, Japan
| | - Shunsuke Aburaya
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hisako Komada
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoko Yamada
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yushi Hirota
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masaru Yoshida
- Department of Food Science and Nutrition, Research Institute of Food and Nutritional Sciences, Graduate School of Human Science and Environment, University of Hyogo, Hyogo, Japan
| | - Munenobu Nogami
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
- Division of Medical Imaging, Biomedical Imaging Research Center, University of Fukui, Fukui, Japan
| | - Takamichi Murakami
- Department of Radiology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Department of Internal Medicine, Kobe University Graduate School of Medicine, Kobe, Japan.
| |
Collapse
|
20
|
Sabatino L, Vassalle C. Thyroid Hormones and Metabolism Regulation: Which Role on Brown Adipose Tissue and Browning Process? Biomolecules 2025; 15:361. [PMID: 40149897 PMCID: PMC11940499 DOI: 10.3390/biom15030361] [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: 02/03/2025] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Thyroid hormones (THs) are important modulators of many metabolic processes, being strictly associated with the control of energy balance, mainly through activities on the brain, white and brown adipose tissue, skeletal muscle, liver, and pancreas. In this review, the principal mechanisms of TH regulation on metabolic processes will be discussed and THs' relevance in metabolic disease progression will be evaluated, especially in the cardiovascular context and correlated diseases. Moreover, we will discuss THs' regulatory role on metabolic events in white and brown adipose tissue, with a special focus on the process of "browning", which consists of the gradual acquisition by white adipocytes of the physical and functional characteristics of brown adipocytes. The advancements in research on molecular mechanisms and proposed physiopathological relevance of this process will be discussed.
Collapse
Affiliation(s)
- Laura Sabatino
- Institute of Clinical Physiology, National Council of Research, 56124 Pisa, Italy
| | | |
Collapse
|
21
|
Bonfante ILP, Segantim HDS, Mendonça KNS, de Oliveira MAB, Monfort-Pires M, Duft RG, da Silva Mateus KC, Chacon-Mikahil MPT, Ramos CD, Velloso LA, Cavaglieri CR. Better cardiometabolic/inflammatory profile is associated with differences in the supraclavicular adipose tissue activity of individuals with T2DM. Endocrine 2025; 87:1011-1021. [PMID: 39627400 DOI: 10.1007/s12020-024-04122-6] [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: 05/03/2024] [Accepted: 11/25/2024] [Indexed: 02/22/2025]
Abstract
PURPOSE Brown adipose tissue (BAT), located in the supraclavicular region, has been associated with a better cardiometabolic profile and reduced risk of developing non-communicable chronic diseases (NCD), in addition to being associated with a healthier phenotype in obesity. However, it is unknown whether greater supraclavicular adipose tissue activity could be associated with a healthier metabolic profile in people already diagnosed with type 2 diabetes (T2DM). Thus, the present work evaluated if supraclavicular adipose tissue activity is associated with metabolic and molecular markers in individuals with T2DM. METHODS Based on a cluster study, individuals with T2DM were divided into groups according to high or low-standard uptake value (SUV) evaluated in the supraclavicular adipose tissue area by [18F]-fluorodeoxyglucose and positron emission tomography-computed tomography (18F-FDG-PET/CT) after mild cold exposure). Functional, biochemical, inflammatory, and molecular markers were measured. RESULTS When we evaluated the whole sample, women showed higher SUV, which favored a difference between groups in sex-related markers. On the other hand, volunteers in the high-SUV group showed lower BMI, monocytes count, triglycerides/glucose index (TYG-index) and z score of metabolic syndrome (MS) values, as well as lower triglycerides, and VLDL concentrations. Moreover, they also had enhanced expression of thermogenic genes in subcutaneous fat. When analyzing only women, the differences in markers associated with sex disappear, and a lower count of leukocytes, platelets, along with lower TYG-index, z score of MS values, and triglycerides, VLDL, LDL, and TNFα concentrations were observed in women with the high SUV. In addition, higher expression of thermogenic genes and BECN1 were detected. CONCLUSION Higher supraclavicular adipose tissue SUV in individuals with T2DM is associated with a better cardiometabolic/inflammatory profile and expression of thermogenic genes. CLINICAL TRIAL REGISTRATION UTN: U1111-1202-1476 - 08/20/2020.
Collapse
Affiliation(s)
- Ivan Luiz Padilha Bonfante
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil.
- Postdoctoral Researcher Program (PPPD), University of Campinas, Campinas, Brazil.
| | - Higor da Silva Segantim
- Higher Interdisciplinary Training Program (PROFIS), University of Campinas, Campinas, SP, Brazil
| | | | | | - Milena Monfort-Pires
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, Brazil
- Turku Pet Centre, University of Turku, Turku, Finland
| | - Renata Garbellini Duft
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Ashgrove Rd W, Aberdeen, Scotland, UK
| | | | | | - Celso Darío Ramos
- Department of Radiology, University of Campinas, Campinas, SP, Brazil
| | - Licio Augusto Velloso
- Laboratory of Cell Signaling, Department of Internal Medicine, University of Campinas, Campinas, SP, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, Brazil
| | - Cláudia Regina Cavaglieri
- Laboratory of Exercise Physiology, School of Physical Education, University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
22
|
Ishida Y, Nakayama K. Infrared thermography unveiled the variation of brown adipose tissue thermogenesis among East Asian adults. Physiol Rep 2025; 13:e70279. [PMID: 40110933 PMCID: PMC11923896 DOI: 10.14814/phy2.70279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/21/2025] [Accepted: 03/07/2025] [Indexed: 03/22/2025] Open
Abstract
The thermogenesis of brown adipose tissue (BAT) is interesting because the contribution to human adaptation to cold and obesity resistance has been suggested. 18F-fluorodeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) is a common method for measuring BAT activity; however, it has been studied in few large cohorts due to concerns about safety and cost. Studies using alternative methods make it challenging to directly compare BAT activity among studies and interpret those results because the procedure is various. We measured the supraclavicular BAT thermogenesis of 122 healthy Japanese and Chinese adults under mild cold stress using standardized infrared thermography (IRT) and examined the effects of various factors on BAT variation. BAT thermogenesis was significantly higher in females than in males (p < 0.001) and significantly higher in Chinese than in Japanese individuals (p < 0.05). Among the 27 participants enrolled in both summer and winter experiments, BAT thermogenesis increased during winter (p < 0.05) only in Japanese participants. Additionally, individuals born at higher latitudes exhibited greater BAT thermogenesis (p < 0.05), suggesting the involvement of genetic background or cold exposure in early life stages. We obtained interesting anthropological and physiological findings with the use of non-invasive IRT.
Collapse
Affiliation(s)
- Yuka Ishida
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Kazuhiro Nakayama
- Department of Integrated Biosciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| |
Collapse
|
23
|
Pahlavani M, Pham K, Kalupahana NS, Morovati A, Ramalingam L, Abidi H, Kiridana V, Moustaid-Moussa N. Thermogenic adipose tissues: Promising therapeutic targets for metabolic diseases. J Nutr Biochem 2025; 137:109832. [PMID: 39653156 DOI: 10.1016/j.jnutbio.2024.109832] [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: 05/02/2024] [Revised: 11/30/2024] [Accepted: 12/04/2024] [Indexed: 01/03/2025]
Abstract
The ongoing increase in the prevalence of obesity and its comorbidities such as cardiovascular disease, type 2 diabetes (T2D) and dyslipidemia warrants discovery of novel therapeutic options for these metabolic diseases. Obesity is characterized by white adipose tissue expansion due to chronic positive energy balance as a result of excessive energy intake and/or reduced energy expenditure. Despite various efforts to prevent or reduce obesity including lifestyle and behavioral interventions, surgical weight reduction approaches and pharmacological methods, there has been limited success in significantly reducing obesity prevalence. Recent research has shown that thermogenic adipocyte (brown and beige) activation or formation, respectively, could potentially act as a therapeutic strategy to ameliorate obesity and its related disorders. This can be achieved through the ability of these thermogenic cells to enhance energy expenditure and regulate circulating levels of glucose and lipids. Thus, unraveling the molecular mechanisms behind the formation and activation of brown and beige adipocytes holds the potential for probable therapeutic paths to combat obesity. In this review, we provide a comprehensive update on the development and regulation of different adipose tissue types. We also emphasize recent interventions in harnessing therapeutic potential of thermogenic adipocytes by bioactive compounds and new pharmacological anti-obesity agents.
Collapse
Affiliation(s)
- Mandana Pahlavani
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Sciences, Texas Woman's University, Dallas, Texas, USA
| | - Kenneth Pham
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, UAE
| | - Ashti Morovati
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Department of Nutrition and Food Studies, Syracuse University, Syracuse, New York, USA
| | - Hussain Abidi
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA
| | - Vasana Kiridana
- Faculty of Medicine, University of Peradeniya, Peradeniya, Sri Lanka
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, Texas, USA; Obesity Research Institute, Texas Tech University, Lubbock, Texas, USA; Institute for One Health Innovation, Texas Tech University and Texas Tech Health Sciences Center, Lubbock, Texas, USA.
| |
Collapse
|
24
|
Zhang Y, Zhou S, Zhao R, Xiong C, Huang Y, Zhang M, Wang Y. Multilayer regulation of postprandial triglyceride metabolism in response to acute cold exposure. J Lipid Res 2025; 66:100751. [PMID: 39892721 PMCID: PMC11903801 DOI: 10.1016/j.jlr.2025.100751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/22/2025] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
Triglyceride-rich lipoproteins carry lipids in the bloodstream, where the fatty acid moieties are liberated by lipoprotein lipase (LPL) and taken up by peripheral tissues such as brown adipose tissue (BAT) and white adipose tissue (WAT), whereas the remaining cholesterol-rich remnant particles are cleared mainly by the liver. Elevated triglyceride (TG) levels and prolonged circulation of cholesterol-rich remnants are risk factors for cardiovascular diseases. Acute cold exposure decreases postprandial TG levels and is a potential therapeutic approach to treat hypertriglyceridemia. However, how acute cold exposure regulates TG metabolism remains incompletely understood. In the current study, we found that acute cold exposure simultaneously increases postprandial very-low-density lipoprotein production and TG clearance, with the latter playing a dominant role and resulting in decreased TG levels. Acute cold exposure increases LPL activity and TG uptake in BAT, while suppressing LPL activity and TG uptake in WAT. Mechanistically, acute cold exposure increases BAT LPL activity through transcriptional upregulation of Lpl and posttranscriptional regulation via inhibiting the hepatic insulin-ANGPTL8-ANGPTL3 axis, while suppressing WAT LPL activity through upregulation of ANGPTL4. Angptl8 knockout mice have dramatically decreased levels of circulating TG. In the absence of ANGPTL8, acute cold exposure increases rather than decreases circulating TG levels. Thus, our study reveals multilayered regulation of acute cold response and postprandial TG metabolism, highlighting the key functions of ANGPTL3, 4, and 8 in response to acute cold exposure.
Collapse
Affiliation(s)
- Yiliang Zhang
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Shengyang Zhou
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Runming Zhao
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Chunyu Xiong
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yingzhen Huang
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Minzhu Zhang
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
| | - Yan Wang
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
25
|
Wang S, He T, Luo Y, Ren K, Shen H, Hou L, Wei Y, Fu T, Xie W, Wang P, Hu J, Zhu Y, Huang Z, Li Q, Li W, Guo H, Li B. SOX4 facilitates brown fat development and maintenance through EBF2-mediated thermogenic gene program in mice. Cell Death Differ 2025; 32:447-465. [PMID: 39402212 PMCID: PMC11893884 DOI: 10.1038/s41418-024-01397-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 03/12/2025] Open
Abstract
Brown adipose tissue (BAT) is critical for non-shivering thermogenesis making it a promising therapeutic strategy to combat obesity and metabolic disease. However, the regulatory mechanisms underlying brown fat formation remain incompletely understood. Here, we found SOX4 is required for BAT development and thermogenic program. Depletion of SOX4 in BAT progenitors (Sox4-MKO) or brown adipocytes (Sox4-BKO) resulted in whitened BAT and hypothermia upon acute cold exposure. The reduced thermogenic capacity of Sox4-MKO mice increases their susceptibility to diet-induced obesity. Conversely, overexpression of SOX4 in BAT enhances thermogenesis counteracting diet-induced obesity. Mechanistically, SOX4 activates the transcription of EBF2, which determines brown fat fate. Moreover, phosphorylation of SOX4 at S235 by PKA facilitates its nuclear translocation and EBF2 transcription. Further, SOX4 cooperates with EBF2 to activate transcriptional programs governing thermogenic gene expression. These results demonstrate that SOX4 serves as an upstream regulator of EBF2, providing valuable insights into BAT development and thermogenic function maintenance.
Collapse
Affiliation(s)
- Shuai Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Ting He
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Ya Luo
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, 361102, Xiamen, Fujian, China
| | - Kexin Ren
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Huanming Shen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Science, 518055, Shenzhen, China
| | - Lingfeng Hou
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yixin Wei
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Tong Fu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Wenlong Xie
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Peng Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Jie Hu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Yu Zhu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China
| | - Zhengrong Huang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China
| | - Qiyuan Li
- National Institute for Data Science in Health and Medicine, School of Medicine, Xiamen University, 361102, Xiamen, Fujian, China.
| | - Weihua Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, 361102, Xiamen, China.
| | - Huiling Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
| | - Boan Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network and Engineering Research Center of Molecular Diagnostics of The Ministry of Education, School of Life Sciences, Xiamen University, 361102, Xiamen, Fujian, China.
| |
Collapse
|
26
|
Guan H, Wang L, Tang C, Xu H, Xiang A, Chen X, Yu Q, Xu L. IDOL alleviates the body weight by upregulating UCP-1 in mice. Diabetes Obes Metab 2025; 27:1314-1326. [PMID: 39748219 DOI: 10.1111/dom.16127] [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: 08/08/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND Given the potential role of brown adipose tissue (BAT) in stimulating energy expenditure, activating BAT can be an effective anti-obesity treatment. Here, we aimed to use adenoviruses to establish the effect of the inducible degrader of the low density lipoprotein receptor (IDOL) in the formation of BAT. METHODS IDOL or green fluorescent protein was overexpressed by adenovirus and injected into the scapula of C57BL/6J mice and fed with high-fat diet for 12 weeks. We measured the body weight, morphology of lipid droplets, lipid profiles and adipogenesis protein expression levels. BAT was isolated, and RNA sequencing was performed to identify the differentially expressed genes and related signaling pathways. Finally, we conducted western blot to verify the authenticity and reliability of the RNA sequencing results. RESULTS Compared with the control group, IDOL overexpression led to a significant reduction in body weight, consistent with the weight of adipose tissues and organs. Further studies show IDOL promotion increased ATGL, perilipin 1 and UCP-1 expression in BAT. However, perilipin 1 protein expression was significantly reduced in the Ad-IDOL group in epididymal white adipose tissue, while there was no significant difference in adiponectin, ATGL and perilipin 1 protein expression in inguinal white adipose tissue. Notably, serum FGF21 and leptin protein expression were negatively related to the adipose tissue decrease after Ad-IDOL administration. RNA sequencing analysis identified 1256 differentially expressed genes that were prominently enriched across nine signalling pathways. Additionally, the protein expression of PGAM2, G6PC1 and phosphorylation-AMPK was significantly increased after overexpression IDOL in BAT, which was consistent with the results of the RNA sequencing analysis. CONCLUSIONS Our research demonstrated that IDOL overexpression alleviates the body weight by promoting the phosphorylation of AMPK to upregulate the UCP-1 and ATGL exacerbating lipolysis in BAT.
Collapse
Affiliation(s)
- Hua Guan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Le Wang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Chengcheng Tang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Hao Xu
- Department of Anatomy, Xi'an Medical University, Xi'an, China
| | - Aoqi Xiang
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Xiaochang Chen
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Qi Yu
- Shaanxi Key Laboratory of Ischemic Cardiovascular Diseases & Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Lixian Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Anesthesiology, School of Stomatology, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|
27
|
Shen Z, Tian K, Tang J, Wang L, Zhang F, Yang L, Ge Y, Jiang M, Zhao X, Yang J, Chen G, Wang X. Exposure to Nanoplastics During Pregnancy Induces Brown Adipose Tissue Whitening in Male Offspring. TOXICS 2025; 13:171. [PMID: 40137498 PMCID: PMC11945425 DOI: 10.3390/toxics13030171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/23/2025] [Accepted: 02/26/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Polystyrene nanoplastics (PSNPs) have been recognized as emerging environmental pollutants with potential health impacts, particularly on metabolic disorders. However, the mechanism by which gestational exposure to PSNPs induces obesity in offspring remains unclear. This study, focused on the whitening of brown adipose tissue (BAT), aims to elucidate the fundamental mechanisms by which prenatal exposure to PSNPs promotes obesity development in mouse offspring. METHODS AND RESULTS Pregnant dams were subjected to various doses of PSNPs (0 µg/µL, 0.5 µg/µL, and 1 µg/µL), and their offspring were analyzed for alterations in body weight, adipose tissue morphology, thermogenesis, adipogenesis, and lipophagy. The findings revealed a notable reduction in birth weight and an increase in white adipocyte size in adult offspring mice. Notably, adult male mice exhibited BAT whitening, correlated with a negative dose-dependent downregulation of UCP1 expression, indicating thermogenesis dysfunction. Further investigation revealed augmented lipogenesis evidenced by the upregulation of FASN, SREBP-1c, CD36, and DGAT2 expression, coupled with the inhibition of lipophagy, indicated by elevated levels of mTOR, AKT, and p62 proteins and reduced levels of LC3II/LCI and Lamp2 proteins in male offspring. CONCLUSIONS These findings indicate that gestational PSNP exposure plays a role in the development of obesity in offspring through the whitening of brown adipose tissue, which is triggered by lipogenesis and lipophagy inhibition, providing a novel insight into the metabolic risks associated with gestational PSNPs exposure.
Collapse
Affiliation(s)
- Zhaoping Shen
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Kai Tian
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Jiayi Tang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Lin Wang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Fangsicheng Zhang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Lingjuan Yang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Yufei Ge
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Mengna Jiang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Xinyuan Zhao
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| | - Jinxian Yang
- Xinglin College, Nantong University, Qidong 226236, China;
| | - Guangdi Chen
- Department of Public Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoke Wang
- Nantong Key Laboratory of Environmental Toxicology, Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University, Nantong 226019, China; (Z.S.); (K.T.); (J.T.); (L.W.); (F.Z.); (L.Y.); (Y.G.); (M.J.); (X.Z.)
| |
Collapse
|
28
|
Malmon S, Casasnovas O, Fournier M, Cartron G, Kanoun S, Cottereau AS, Herbaux C, Al Tabaa Y. Personalized baseline and residual TMTV influence treatment response and outcomes in relapsed/refractory lymphomas: results from the GATA study. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07154-w. [PMID: 39984744 DOI: 10.1007/s00259-025-07154-w] [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: 12/18/2024] [Accepted: 02/12/2025] [Indexed: 02/23/2025]
Abstract
PURPOSE Total metabolic tumor volume (TMTV) at baseline becomes a key biomarker in several lymphoma subtypes. Variability in segmentation methods such as 41%SUVmax and SUVmax > 4 has limited its clinical application. Additionally, immune-checkpoint-inhibitors introduced challenges in response assessment due to pseudoprogression, complicating the use of traditional metrics. This study investigates the prognostic impact of baseline- and residual-TMTV and introduces a novel personalized-liver-based-threshold (pTMTVliver) to enhance precision in patient stratification. METHODS We analyzed 91 patients with relapsed/refractory diffuse-large-B-cell lymphoma and follicular lymphoma from the GATA trial, comparing patient's outcome according to three segmentation methods: TMTV41%, TMTV4, and pTMTVliver. pTMTVliver used a threshold of 200%SUVmeanliver aligning with 125%SUVmaxliver to enhance standardization and reduce variability. RESULTS Baseline-TMTV significantly influenced prognosis with higher TMTV correlating with shorter PFS and OS (p < 0.0001 for all methods). Residual-TMTV, particularly with pTMTVliver and TMTV4, stratified no-CMR patients with the lowest predictive errors and better predictive accuracy compared to TMTV41% Multivariate analyses confirmed residual-pTMTVliver as superior for prognostic performance for PFS (HR:5.10; C-index:0.724) and OS (HR:4.00; C-index:0.853) compared to TMTV4 and Deauville Score (DS). The DS alone did not fully capture the heterogeneity of outcomes of DS4-5 patients. CONCLUSION Baseline- and residual-TMTV strongly influence prognosis and response in lymphoma patients. The novel personalized pTMTVliver method offers improved accuracy of patient stratification, particularly for those with DS4-5, providing more reliable risk assessment. Larger cohorts are needed to validate these findings and optimize residual-TMTV-based clinical applications.
Collapse
Affiliation(s)
| | - Olivier Casasnovas
- Department of Hematology, Dijon Bourgogne University Hospital and INSERM UMR 1231, Dijon, France
| | - Marguerite Fournier
- Department of Biostatistics, LYSARC, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Guillaume Cartron
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
| | - Salim Kanoun
- Cancer Research Center of Toulouse, Team 9, INSERM Unité Mixte de Recherche 1037, Toulouse, France
| | - Anne Ségolène Cottereau
- Department of Nuclear Medicine, Cochin Hospital, AP-HP, University of Paris Cité, Paris, France
| | - Charles Herbaux
- Department of Clinical Hematology, CHU Montpellier, Montpellier, France
- Institute of Human Genetics, UMR CNRS-UM, Montpellier, 9002, France
| | - Yassine Al Tabaa
- Scintidoc Nuclear Medicine Center, Clinique Clémentville, Montpellier, France.
| |
Collapse
|
29
|
Wang H, Zhang L, Chen X, Hong L, Zhao J, Qian W, Pham LK, Willard B, Li X, Bulek K, Li X. Adipocyte-specific Steap4 deficiency reduced thermogenesis and energy expenditure in mice. iScience 2025; 28:111903. [PMID: 39995871 PMCID: PMC11848796 DOI: 10.1016/j.isci.2025.111903] [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: 01/22/2024] [Revised: 11/21/2024] [Accepted: 01/22/2025] [Indexed: 02/26/2025] Open
Abstract
Six-transmembrane protein of prostate 4 (Steap4), highly expressed in adipose tissue, is associated with metabolic homeostasis. Dysregulated adipose and mitochondrial metabolism contributes to obesity, highlighting the need to understand their interplay. Whether and how Steap4 influences mitochondrial function, adipocytes, and energy expenditure remain unclear. Adipocyte-specific Steap4-deficient mice exhibited increased fat mass and severe insulin resistance in our high-fat diet model. Mass spectrometry identified two classes of Steap4 interactomes: mitochondrial proteins and proteins involved in splicing. RNA sequencing (RNA-seq) analysis of white adipose tissue demonstrated that Steap4 deficiency altered RNA splicing patterns with enriched mitochondrial functions. Indeed, Steap4 deficiency impaired respiratory chain complex activity, causing mitochondrial dysfunction in white adipose tissue. Consistently, brown adipocyte-specific Steap4 deficiency impaired mitochondrial function, increased brown fat whitening, reduced energy expenditure, and exacerbated insulin resistance in a high-fat model. Overall, our study highlights Steap4's critical role in modulating adipocyte mitochondrial function, thereby controlling thermogenesis, energy expenditure, and adiposity.
Collapse
Affiliation(s)
- Han Wang
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Lizi Zhang
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Xing Chen
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Lingzi Hong
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Junjie Zhao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Wen Qian
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Lam Khue Pham
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Belinda Willard
- Proteomics and Metabolomics Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Xiaoxia Li
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Katarzyna Bulek
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA
| | - Xiao Li
- Department of Biochemistry, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Center for RNA Science and Therapeutics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Computer and Data Sciences, School of Engineering, Case Western Reserve University, Cleveland,OH 44106, USA
| |
Collapse
|
30
|
Das A, Mund C, Hagag E, Garcia-Martin R, Karadima E, Witt A, Peitzsch M, Deussen A, Chavakis T, Noll T, Alexaki VI. Adenylate cyclase 10 promotes brown adipose tissue thermogenesis. iScience 2025; 28:111833. [PMID: 39949963 PMCID: PMC11821413 DOI: 10.1016/j.isci.2025.111833] [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: 05/02/2024] [Revised: 11/01/2024] [Accepted: 01/15/2025] [Indexed: 02/16/2025] Open
Abstract
Brown adipose tissue (BAT) thermogenesis dissipates energy through heat production and thereby it opposes metabolic disease. It is mediated by mitochondrial membrane uncoupling, yet the mechanisms sustaining the mitochondrial membrane potential (ΔΨm) in brown adipocytes are poorly understood. Here we show that isocitrate dehydrogenase (IDH) activity and the expression of the soluble adenylate cyclase 10 (ADCY10), a CO2/bicarbonate sensor residing in mitochondria, are upregulated in BAT of cold-exposed mice. IDH inhibition or ADCY10 deficiency reduces cold resistance of mice. Mechanistically, IDH increases the ΔΨm in brown adipocytes via ADCY10. ADCY10 sustains complex I activity and the ΔΨm via exchange protein activated by cAMP1 (EPAC1). However, neither IDH nor ADCY10 inhibition affect uncoupling protein 1 (UCP1) expression. Hence, we suggest that ADCY10, acting as a CO2/bicarbonate sensor, mediates the effect of IDH on complex I activity through cAMP-EPAC1 signaling, thereby maintaining the ΔΨm and enabling thermogenesis in brown adipocytes.
Collapse
Affiliation(s)
- Anupam Das
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Christine Mund
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eman Hagag
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Ruben Garcia-Martin
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Eleftheria Karadima
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Anke Witt
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mirko Peitzsch
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden of the Helmholtz Center Munich, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Thomas Noll
- Department of Physiology, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Vasileia Ismini Alexaki
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
31
|
Zhao J, Li X, Liang C, Yan Y. Can Exercise-Mediated Adipose Browning Provide an Alternative Explanation for the Obesity Paradox? Int J Mol Sci 2025; 26:1790. [PMID: 40076419 PMCID: PMC11898606 DOI: 10.3390/ijms26051790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 02/06/2025] [Accepted: 02/14/2025] [Indexed: 03/14/2025] Open
Abstract
Overweight patients with cardiovascular disease (CVD) tend to survive longer than normal-weight patients, a phenomenon known as the "obesity paradox". The phenotypic characteristics of adipose distribution in these patients (who survive longer) often reveal a larger proportion of subcutaneous white adipose tissue (scWAT), suggesting that the presence of scWAT is negatively associated with all-cause mortality and that scWAT appears to provide protective benefits in patients facing unhealthy states. Exercise-mediated browning is a crucial aspect of the benign remodeling process of adipose tissue (AT). Reduced accumulation, reduced inflammation, and associated adipokine secretion are directly related to the reduction in CVD mortality. This paper summarized the pathogenetic factors associated with AT accumulation in patients with CVD and analyzed the possible role and pathway of exercise-mediated adipose browning in reducing the risk of CVD and CVD-related mortality. It is suggested that exercise-mediated browning may provide a new perspective on the "obesity paradox"; that is, overweight CVD patients who have more scWAT may gain greater cardiovascular health benefits through exercise.
Collapse
Affiliation(s)
- Jiani Zhao
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
| | - Xuehan Li
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
| | - Chunyu Liang
- School of Physical Education, Guangxi University (GXU), Nanning 530004, China
| | - Yi Yan
- Department of Sport Biochemistry, School of Sport Science, Beijing Sport University (BSU), Beijing 100084, China; (J.Z.); (X.L.)
- Laboratory of Sports Stress and Adaptation of General Administration of Sport, Beijing Sport University (BSU), Beijing 100084, China
- Exercise and Physical Fitness, Beijing Sport University (BSU), Beijing 100084, China
| |
Collapse
|
32
|
Wakabayashi H, Sakaue H, Nishimura T. Recent updates on cold adaptation in population and laboratory studies, including cross-adaptation with nonthermal factors. J Physiol Anthropol 2025; 44:7. [PMID: 39972479 PMCID: PMC11837704 DOI: 10.1186/s40101-025-00387-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
This review aims to update our understanding of human cold adaptation. First, an overview of the thermoregulatory response to cold is provided, with some recent updates in human brown adipose tissue (BAT). Variation in BAT activity and multiorgan contributions to cold-induced thermogenesis were introduced. We found that individuals with less BAT activity rely more on shivering to compensate for less non-shivering thermogenesis (NST). The mechanisms of cold-induced vasoconstriction are summarized, including the role of arteriovenous anastomoses, adrenergic neural function, and inhibition of the nitric oxide vasodilator pathway. In addition, cold-induced vasodilation (CIVD) during cold immersion of the distal extremities is summarized with some recent updates in physiological mechanism. Furthermore, the cold shock response at the onset of cold immersion is introduced. Next, categorization of cold acclimatization/acclimation into habituation of shivering and metabolic and insulative adaptation are provided, with some recent updates. Especially, the rediscovery of human BAT has clarified metabolic acclimation, where increased NST replace shivering. Then, a greater CIVD response in populations in cold regions has been reported, whereas recent laboratory studies suggest no increase in CIVD after repeated cold exposure. To prevent cold injuries, individuals should not rely on habituation through repeated cold exposure. In addition, habituation to the cold shock response after repeated cold water immersion could help reduce the number of drownings. Furthermore, cross-adaptation between cold and nonthermal factors in the thermoregulatory response is summarized. Recent studies explored the relationship between exercise training and BAT activity, although this remains unresolved, depending on the exercise intensity and environmental conditions. The effects of exercise with cold exposure on the thermoregulatory response to cold are summarized in studies including divers working in cold water. We investigated the effect of exercise training in cold water, which resulted in increased muscle deoxygenation during submaximal exercise and greater anerobic power. Moreover, the effects of a hypoxic environment on cold adaptation are summarized. Elevated basal metabolism and higher distal skin temperature in highlanders could improve their cold tolerance. Finally, factors affecting cold adaptation are discussed. The type of cold adaptation may depend on the specific thermoregulatory responses repeated during the adaptation process.
Collapse
Affiliation(s)
- Hitoshi Wakabayashi
- Faculty of Engineering, Hokkaido University, N13 W8 Kita-Ku, Sapporo, 060-8628, Japan
| | - Hiroyuki Sakaue
- Faculty of Health and Sport Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba, 305-8574, Japan
| | - Takayuki Nishimura
- Faculty of Design, Kyushu University, 4-9-1 Shiobaru, Minami-Ku, Fukuoka, 815-8540, Japan.
| |
Collapse
|
33
|
Feng L, Cui J, Chen W, Zhu L, Li P, Zhou H, Sun Y, Yi W. Nrg4 Secreted by Brown Adipose Tissue Suppresses Ferroptosis of Sepsis-Induced Liver Injury. Inflammation 2025:10.1007/s10753-024-02230-z. [PMID: 39956880 DOI: 10.1007/s10753-024-02230-z] [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: 10/16/2024] [Revised: 12/07/2024] [Accepted: 12/23/2024] [Indexed: 02/18/2025]
Abstract
Sepsis is a leading cause of death, with the liver being particularly vulnerable to sepsis-related injuries. This damage significantly contributes to disease progression, underscoring the need for new treatments. Brown adipose tissue (BAT) secretes various cytokines, including neuregulin 4 (Nrg4), which plays a protective role in hepatic glucose and lipid metabolism. Ferroptosis, a key type of cell death in sepsis-induced liver injury, has recently gained attention. This study aimed to investigate how BAT-secreted cytokines alleviate liver ferroptosis in sepsis. Septic liver injury was induced in the control and BAT group using cecal ligation and puncture (CLP) and lipopolysaccharide injections. BAT removal worsened ferroptosis; in contrast, CL316243 activation reduced it. These findings suggest that Nrg4 secretion following BAT activation protects the liver during sepsis by inhibiting ferroptosis. Future therapies targeting BAT activation and Nrg4 could potentially mitigate sepsis-induced liver damage, offering new insights into treatment strategies.
Collapse
Affiliation(s)
- Linqi Feng
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun Cui
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Wenlong Chen
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Zhu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Panpan Li
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Haitao Zhou
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yang Sun
- Department of Geriatrics, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| | - Wei Yi
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
34
|
Batrow PL, Roux CH, Gautier N, Martin L, Sibille B, Guillou H, Postic C, Langin D, Mothe-Satney I, Amri EZ. Regulation of UCP1 expression by PPARα and pemafibrate in human beige adipocytes. Life Sci 2025; 363:123406. [PMID: 39828228 DOI: 10.1016/j.lfs.2025.123406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
AIMS Thermogenic adipocytes are able to dissipate energy as heat from lipids and carbohydrates through enhanced uncoupled respiration, due to UCP1 activity. PPAR family of transcription factors plays an important role in adipocyte biology. The purpose of this work was to characterize the role of PPARα and pemafibrate in the control of thermogenic adipocyte formation and function. MATERIALS AND METHODS We used human multipotent adipose-derived stem cells and primary cultures of stroma-vascular fraction cells, transfected with siRNA against PPARα, differentiated into white or beige adipocytes, by the treatment of rosiglitazone or pemafibrate. The expression of key marker genes of adipogenesis and thermogenesis was determined using RT-qPCR and Western blotting. An RNAseq analysis was also performed. KEY FINDINGS We show that inhibition of PPARα mRNA increases UCP1 mRNA and protein expression in beige adipocytes induced by rosiglitazone. Knock-down of PPARα also increases stimulated glycerol release. Pemafibrate, described as a selective PPARα modulator, induces adipogenesis and the expression of UCP1 in the absence of PPARα expression. These effects are inhibited by a specific PPARγ antagonist highly suggesting that the pemafibrate effects in adipogenesis and beiging were mediated by PPARγ. SIGNIFICANCE Conversion of white into thermogenic adipocytes is mainly due to the activation of PPARγ. Moreover, we show that PPARα seems to act as a hindrance for PPARγ-dependent beiging. Our data question the role of PPARα in human adipocyte browning and the specificity of pemafibrate in adipocytes.
Collapse
Affiliation(s)
- Pierre-Louis Batrow
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France
| | - Christian H Roux
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France; Rheumatology Department, Hospital Pasteur 2 CHU, Adipo-Cible Research Study Group, Nice, France
| | - Nadine Gautier
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France
| | - Luc Martin
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France
| | - Brigitte Sibille
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France
| | - Hervé Guillou
- Toxalim (Research Centre in Food Toxicology), université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Catherine Postic
- Université Paris Cité, Institut Cochin, CNRS, Inserm, Paris, France
| | - Dominique Langin
- Institute of Metabolic and Cardiovascular Diseases, I2MC, University of Toulouse, Inserm, Toulouse III University - Paul Sabatier (UPS), Toulouse, France; Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Institut Universitaire de France (IUF), Paris, France
| | - Isabelle Mothe-Satney
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France
| | - Ez-Zoubir Amri
- Université Côte d'Azur, CNRS, Inserm, Adipo-Cible Research Study Group, iBV, Nice, France.
| |
Collapse
|
35
|
Reyad-Ul Ferdous M, Wolde T, Pandey V, Qin P. Mitochondrial function, cell viability, pharmacokinetics and molecular simulation studies reveal the impact of CX-6258 HCl, a pan-Pim kinase inhibitor, on adipocytes. J Biomol Struct Dyn 2025:1-13. [PMID: 39936172 DOI: 10.1080/07391102.2025.2460738] [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: 11/28/2023] [Accepted: 12/15/2024] [Indexed: 02/13/2025]
Abstract
Obesity leads to the development of several diseases and chronic death worldwide. Mitochondrial dysfunction is one of the vital causes to develop obesity. Targeting mitochondrial uncoupling protein 1 (UCP1) may well be a potential therapeutic approach against obesity or mitochondrial dysfunction-related illnesses. To assess the significance of mitochondrial adenosine triphosphate (ATP) synthesis, mitochondrial DNA quantity and in vitro pharmacodynamics and pharmacokinetics, we used CX-6258 HCl (pan-Pim kinase inhibitor) in this work. CX-6258 HCl significantly reduces ATP production both in white and brown adipocytes and, therefore, improves thermogenesis, which helps to reduce fat in adipocytes. On the HEK293T cell line, no appreciable cell growth was seen. The in silico analysis identifies a potential interaction between CX-6258 HCl and the UCP1 protein. To treat disorders linked to mitochondrial dysfunction or obesity, CX-6258 HCl may be a promising therapeutic option. The role of pan-Pim kinase inhibitor on obesity and mitochondrial dysfunction-related disorders remains unknown. Further investigation will be leading to the development of the mechanism of action and therapeutic potential of CX-6258 HCl (pan-Pim kinase inhibitor).
Collapse
Affiliation(s)
- Md Reyad-Ul Ferdous
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| | - Tesfaye Wolde
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| | - Vijay Pandey
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| | - Peiwu Qin
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, PR China
| |
Collapse
|
36
|
Kong G, Koh J, Chia J, Neo B, Chen Y, Cao G, Chong B, Muthiah M, Sim HW, Ng G, Koo CY, Khoo CM, Chan MYY, Loh PH, Chew NWS. A sex-disaggregated analysis of the prognostic value of lean type 2 diabetes mellitus in the adult population with acute myocardial infarction. Cardiovasc Diabetol 2025; 24:59. [PMID: 39920748 PMCID: PMC11806904 DOI: 10.1186/s12933-024-02552-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Emerging evidence has demonstrated the unfavourable cardiovascular risk of individuals with lean type 2 diabetes mellitus (T2DM). Our study aims to investigate the prognostic value of lean T2DM in patients with acute myocardial infarction (AMI), stratified by sex. METHODS The study cohort examines the clinical characteristics and long-term outcomes of individuals with AMI, stratified by four phenotypes based on T2DM and lean body category-lean T2DM, non-lean T2DM, lean non-T2DM and non-lean non-T2DM. The primary outcome was long-term all-cause mortality. Cox regression model was constructed to investigate the associations of lean and non-lean T2DM phenotypes with mortality, adjusted for age, ethnicity, previous AMI, AMI type, chronic kidney disease, angiotensin converting enzyme inhibitor or angiotensin receptor blockers, beta-blockers, and smoking status. RESULTS A cohort of 9545 AMI patients was examined, with a mean follow-up duration of 3.4 ± 2.4 years. Majority had the non-lean T2DM phenotype (40.4%), followed by non-lean non-T2DM (29.8%), lean non-T2DM (15.9%), and lean T2DM (13.9%). In the T2DM group, one-quarter was lean (N = 1324), while the vast majority (74.5%) was non-lean. Individuals with lean T2DM tended to be female and older. Patients with lean T2DM had the highest rates of heart failure (23.3%, p < 0.001), cardiogenic shock (9.1%, p = 0.036), and long-term all-cause mortality (32.6%, p < 0.001). Cox regression demonstrated that lean T2DM was an independent predictor of mortality (adjusted hazard ratio [aHR] 1.171, 95% CI 1.040-1.319, p = 0.009) after adjustment. The presence of higher mortality risk following AMI was present in males (aHR 1.201, 95% CI 1.037-1.391, p = 0.015), but not in females (aHR 1.066, 95% CI 0.869-1.308, p = 0.538). CONCLUSIONS The lean T2DM phenotype was present in one-quarter of the AMI cohort with T2DM. The lean T2DM phenotype was an independent predictor of long-term mortality following AMI, although this association was stronger in males than in females.
Collapse
Affiliation(s)
- Gwyneth Kong
- Department of Medicine, National University Hospital, Singapore, Singapore
| | - Jaycie Koh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jobelle Chia
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
| | - Bryan Neo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yiming Chen
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Grace Cao
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Bryan Chong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mark Muthiah
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, Singapore, Singapore
- National University Centre for Organ Transplantation, National University Health System, Singapore, Singapore
| | - Hui Wen Sim
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Division of Cardiology, Department of Medicine, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Gavin Ng
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chieh Yang Koo
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chin Meng Khoo
- Division of Endocrinology, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Mark Yan-Yee Chan
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Poay-Huan Loh
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Cardiology, Department of Medicine, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Nicholas W S Chew
- Department of Cardiology, National University Heart Centre, National University Health System, Singapore, Singapore.
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
37
|
Alpaslan Ağaçdiken A, Göktaş Z. Berberine-induced browning and energy metabolism: mechanisms and implications. PeerJ 2025; 13:e18924. [PMID: 39931072 PMCID: PMC11809318 DOI: 10.7717/peerj.18924] [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: 10/29/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Obesity has become a global pandemic. The approaches researched to prevent it include decreasing energy intake and/or enhancing energy expenditure. Therefore, research on brown adipose tissue is of great importance. Brown adipose tissue is characterized by its high mitochondrial content. Mitochondrial uncoupling protein 1 (UCP1) releases energy as heat instead of chemical energy. Thermogenesis increases energy expenditure. Berberine, a phytochemical widely used in Asian countries, has positive effects on body weight control. While the precise mechanisms behind this effect remain unclear, the adenosine monophosphate-activated protein kinase (AMPK) pathway is known to play a crucial role. Berberine activates AMPK through phosphorylation, significantly impacting brown adipose tissue by enhancing lipolytic activity and increasing the expression of UCP1, peroxisome proliferator-activated receptor γ-co-activator-1α (PGC1α), and PR domain containing 16 (PRDM16). While investigating the mechanism of action of berberine, both the AMPK pathway is being examined in more detail and alternative pathways are being explored. One such pathway is growth differentiation factor 15 (GDF15), known for its appetite-suppressing effect. Berberine's low stability and bioavailability, which are the main obstacles to its clinical use, have been improved through the development of nanotechnological methods. This review examines the potential mechanisms of berberine on browning and summarizes the methods developed to enhance its effect.
Collapse
Affiliation(s)
| | - Zeynep Göktaş
- Department of Nutrition and Dietetics, Hacettepe University, Ankara, Turkey
| |
Collapse
|
38
|
Malaguarnera M, Cauli O, Cabrera-Pastor A. Obesity and Adipose-Derived Extracellular Vesicles: Implications for Metabolic Regulation and Disease. Biomolecules 2025; 15:231. [PMID: 40001534 PMCID: PMC11853251 DOI: 10.3390/biom15020231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Obesity, a global epidemic, is a major risk factor for chronic diseases such as type 2 diabetes, cardiovascular disorders, and metabolic syndrome. Adipose tissue, once viewed as a passive fat storage site, is now recognized as an active endocrine organ involved in metabolic regulation and inflammation. In obesity, adipose tissue dysfunction disrupts metabolic balance, leading to insulin resistance and increased production of adipose-derived extracellular vesicles (AdEVs). These vesicles play a key role in intercellular communication and contribute to metabolic dysregulation, affecting organs such as the heart, liver, and brain. AdEVs carry bioactive molecules, including microRNAs, which influence inflammation, insulin sensitivity, and tissue remodeling. In the cardiovascular system, AdEVs can promote atherosclerosis and vascular dysfunction, while those derived from brown adipose tissue offer cardioprotective effects. In type 2 diabetes, AdEVs exacerbate insulin resistance and contribute to complications such as diabetic cardiomyopathy and cognitive decline. Additionally, AdEVs are implicated in metabolic liver diseases, including fatty liver disease, by transferring inflammatory molecules and lipotoxic microRNAs to hepatocytes. These findings highlight the role of AdEVs in obesity-related metabolic disorders and their promise as therapeutic targets for related diseases.
Collapse
Affiliation(s)
- Michele Malaguarnera
- Psychobiology Department, University of Valencia, 46010 Valencia, Spain;
- Nursing Department, University of Valencia, 46010 Valencia, Spain
| | - Omar Cauli
- Nursing Department, University of Valencia, 46010 Valencia, Spain
- Frailty Research Organized Group (FROG), University of Valencia, 46010 Valencia, Spain
| | - Andrea Cabrera-Pastor
- Pharmacology Department, University of Valencia, 46010 Valencia, Spain;
- Fundación de Investigación del Hospital Clínico Universitario de Valencia (INCLIVA), 46010 Valencia, Spain
| |
Collapse
|
39
|
Martin-Garcia E, Domingo-Rodriguez L, Lutz B, Maldonado R, Ruiz de Azua I. Cannabinoid type-1 receptors in CaMKII neurons drive impulsivity in pathological eating behavior. Mol Metab 2025; 92:102096. [PMID: 39788291 PMCID: PMC11787564 DOI: 10.1016/j.molmet.2025.102096] [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: 10/27/2024] [Revised: 01/03/2025] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
OBJECTIVES Overconsumption of palatable food and energy accumulation are evolutionary mechanisms of survival when food is scarce. These innate mechanisms becom detrimental in obesogenic environment promoting obesity and related comorbidities, including mood disorders. This study aims at elucidating the role of the endocannabinoid system in energy accumulation and hedonic feeding. METHODS We applied a genetic strategy to reconstitute cannabinoid type-1 receptor (CB1) expression at functional levels specifically in CaMKII+ neurons (CaMKII-CB1-RS) and adipocytes (Ati-CB1-RS), respectively, in a CB1 deficient background. RESULTS Rescued CB1 expression in CaMKII+ neurons, but not in adipocytes, promotes feeding behavior, leading to fasting-induced hyperphagia, increased motivation, and impulsivity to palatable food seeking. In a diet-induced obesity model, CB1 re-expression in CaMKII+ neurons, but not in adipocytes, compared to complete CB1 deficiency, was sufficient to largely restore weight gain, food intake without any effect on glucose intolerance associated with high-fat diet consumption. In a model of glucocorticoid-mediated metabolic syndrome, CaMKII-CB1-RS mice showed all metabolic alterations linked to the human metabolic syndrome except of glucose intolerance. In a binge-eating model mimicking human pathological feeding, CaMKII-CB1-RS mice showed increased seeking and compulsive behavior to palatable food, suggesting crucial roles in foraging and an enhanced susceptibility to addictive-like eating behaviors. Importantly, other contingent behaviors, including increased cognitive flexibility and reduced anxiety-like behaviors, but not depressive-like behaviors, were also observed. CONCLUSIONS CB1 in CaMKII+ neurons is instrumental in feeding behavior and energy storage under physiological conditions. The exposure to risk factors (hypercaloric diet, glucocorticoid dysregulation) leads to obesity, metabolic syndrome, binge-eating and food addiction.
Collapse
Affiliation(s)
- Elena Martin-Garcia
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain; Department of Psychobiology and Methodology in Health Sciences, Universitat Autonoma de Barcelona, 08193, Bellatera, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Laura Domingo-Rodriguez
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain; Department of Psychobiology and Methodology in Health Sciences, Universitat Autonoma de Barcelona, 08193, Bellatera, Spain
| | - Beat Lutz
- Leibniz Institute for Resilience Research, 55122, Mainz, Germany; Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128, Mainz, Germany
| | - Rafael Maldonado
- Laboratory of Neuropharmacology-Neurophar, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003, Barcelona, Spain; Department of Psychobiology and Methodology in Health Sciences, Universitat Autonoma de Barcelona, 08193, Bellatera, Spain; Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Inigo Ruiz de Azua
- Leibniz Institute for Resilience Research, 55122, Mainz, Germany; Institute of Physiological Chemistry, University Medical Center of the Johannes Gutenberg University, 55128, Mainz, Germany.
| |
Collapse
|
40
|
Page B, Cora C, Reilly J, Reno R, Harbi W, Lynes MS, Lynes MA, Lynes MD. Monitoring Mouse Surface Temperature During Stress with a Thermal Camera: A Low-Cost Infrared Videography System for Evaluating Murine Metabolism. Curr Protoc 2025; 5:e70098. [PMID: 39945421 PMCID: PMC11823340 DOI: 10.1002/cpz1.70098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2025]
Abstract
Energy is required for life, and organisms obtain their energy from fuel sources to enable both anabolic and catabolic processes. Some of this energy is radiated as heat, which can be quantified as a measure of metabolic rate. In some cases, environmental toxicants can alter metabolic energy in undesirable ways, and characterization of new pharmaceuticals can determine the efficacy of desirable metabolic rate manipulation or identify off-target adverse effects. Current methods to directly measure heat production in laboratory mice are expensive, can be laborious, and make it challenging to monitor animals in ways that are multiplexed, robust, and non-invasive. We present a set of protocols for assembling and deploying a simple, low-cost thermal camera to monitor and record thermogenic activity, modified from prior work. Parts used to build this system currently cost approximately $150 USD and, when assembled, can record mouse temperatures as well as ambient cage temperatures up to twice per second for extended periods. By using multiplexed cameras in a diurnal mouse incubator system, the thermogenic capacity of several mice can be simultaneously recorded and graphed. Exogenous agents and genotypes that alter metabolism can be readily identified with this technology. In this set of protocols, we describe the assembly of the thermal video camera device, its use, and related data capture and analysis methods. © 2025 The Author(s). Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Assembling thermal camera for thermogenic stress test Basic Protocol 2: In vivo measurement of mouse temperature under different ambient conditions.
Collapse
Affiliation(s)
- Breanna Page
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
- Roux Institute at Northeastern UniversityPortlandMaine
| | - Carolina Cora
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
- Graduate School of Biomedical Science and EngineeringUniversity of MaineOronoMaine
| | - James Reilly
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
| | - Ryan Reno
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
| | - Wadak Harbi
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
| | | | - Michael A. Lynes
- Department of Molecular and Cell BiologyUniversity of ConnecticutStorrsConnecticut
| | - Matthew D. Lynes
- Center for Molecular MedicineMaineHealth Institute for ResearchScarboroughMaine
- Roux Institute at Northeastern UniversityPortlandMaine
- Graduate School of Biomedical Science and EngineeringUniversity of MaineOronoMaine
- Department of MedicineMaineHealthPortlandMaine
| |
Collapse
|
41
|
Yu J, Gu X, Guo Y, Gao M, Cheng S, Meng M, Cui X, Zhang Z, Guo W, Yan D, Sheng M, Zhai L, Ji J, Ma X, Li Y, Cao Y, Wu X, Zhao J, Hu Y, Tan M, Lu Y, Xu L, Liu B, Hu C, Ma X. E3 ligase FBXW7 suppresses brown fat expansion and browning of white fat. EMBO Rep 2025; 26:748-767. [PMID: 39747664 PMCID: PMC11811183 DOI: 10.1038/s44319-024-00337-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 10/26/2024] [Accepted: 11/08/2024] [Indexed: 01/04/2025] Open
Abstract
Thermogenic fat, including brown and beige fat, dissipates heat via thermogenesis and enhances energy expenditure. Thus, its activation represents a therapeutic strategy to combat obesity. Here, we demonstrate that levels of F-box and WD repeat domain-containing 7 (FBXW7), an E3 ubiquitin protein ligase, negatively correlate with thermogenic fat functionality. FBXW7 overexpression in fat suppresses energy expenditure and thermogenesis, thus aggravates obesity and metabolic dysfunctions in mice. Conversely, FBXW7 depletion in fat leads to brown fat expansion and browning of white fat, and protects mice from diet induced obesity, hepatic steatosis, and hyperlipidemia. Mechanistically, FBXW7 binds to S6K1 and promotes its ubiquitination and proteasomal degradation, which in turn impacts glycolysis and brown preadipocyte proliferation via lactate. Besides, the beneficial metabolic effects of FBXW7 depletion in fat are attenuated by fat-specific knockdown of S6K1 in vivo. In summary, we provide evidence that adipose FBXW7 acts as a major regulator for thermogenic fat biology and energy homeostasis and serves as potential therapeutic target for obesity and metabolic diseases.
Collapse
Grants
- 32325024,82300979,32222024,32271224,32071148,22225702,82000802 MOST | National Natural Science Foundation of China (NSFC)
- 2023YFA1800400,2019YFA09004500 MOST | National Key Research and Development Program of China (NKPs)
- 22ZR1421200,21140904300 Science and Technology Commission of Shanghai Municipality (STCSM)
- CSTB2022NSCQ-JQX0033 Natural Science Foundation of Chongqing, China
- 2021C03069 Key Research and Development Project of Zhejiang Province, China
- LY20H070003 Zhejiang Provincial Natural Science Foundation of China
- SHSMU-ZDCX20212700 Innovation research team of high-level local universities in Shanghai
- 2022ZZ01002 Shanghai Research Center for Endocrine and Metabolic Diseases
- 2023M741184 China Postdoctoral Science Foundation(China Postdoctoral Foundation Project)
Collapse
Affiliation(s)
- Jian Yu
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xuejiang Gu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yingying Guo
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
| | - Mingyuan Gao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Shimiao Cheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Meiyao Meng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xiangdi Cui
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhe Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Wenxiu Guo
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Dandan Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
| | - Maozheng Sheng
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Linhui Zhai
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jing Ji
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Xinhui Ma
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China
| | - Yu Li
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yuxiang Cao
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Xia Wu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jiejie Zhao
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200000, China
| | - Yepeng Hu
- Department of Endocrine and Metabolic Diseases, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Minjia Tan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yan Lu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China
- Ministry of Education Key Laboratory of Metabolism and Molecular Medicine, Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai, 200000, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute for Aging, East China Normal University, Shanghai, 200241, China.
| | - Bin Liu
- Jiangsu Key Laboratory of Marine Pharmaceutical Compound Screening, College of Pharmacy, Jiangsu Ocean University, Lianyungang, 222005, China.
| | - Cheng Hu
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China.
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Centre for Diabetes, Shanghai, 200233, China.
| | - Xinran Ma
- Joint Center for Translational Medicine, Fengxian District Central Hospital, Fengxian District, Shanghai, 201400, China.
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Chongqing Key Laboratory of Precision Optics, Chongqing Institute of East China Normal University, Chongqing, 401120, China.
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- Institute for Aging, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
42
|
Hankir MK. Creating a picture of brown fat with creatine-CEST. Trends Endocrinol Metab 2025; 36:102-104. [PMID: 39256118 DOI: 10.1016/j.tem.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
Accurate assessment of brown fat thermogenesis by non-invasive means remains challenging. Writing in Nature Metabolism, Cai et al. leverage the futile creatine cycling characteristic of thermogenic adipocytes to show that a type of magnetic resonance imaging (MRI) technique sensitive to endogenous creatine levels faithfully tracks brown fat thermogenesis in rodents and in humans.
Collapse
Affiliation(s)
- Mohammed K Hankir
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
43
|
Harrison DB, Phillips AL, Tansey JB, Clarke TJ, Wood CB, Nedzi L, Schwartz DL, Makowski L, Hayes DN, Newman G, Gleysteen JP. Brown Adipose Tissue Mimicking Head and Neck Cancer on PET Scan in a Patient on GLP-1 Drug. Laryngoscope 2025; 135:741-743. [PMID: 39370986 PMCID: PMC11729570 DOI: 10.1002/lary.31815] [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: 09/08/2024] [Accepted: 09/17/2024] [Indexed: 10/08/2024]
Abstract
To report a case of a patient undergoing GLP-1 receptor agonist therapy in which increased FDG uptake in brown adipose tissue (BAT) mimicked metastatic head and neck cancer on PET/CT imaging. A 61-year-old female with Class III obesity presented with a right-sided neck mass after significant weight loss following the use of the GLP-1 receptor agonist, Semaglutide. PET/CT revealed FDG uptake in the right level II lymph node and extensive BAT uptake throughout the neck and mediastinum, complicating the diagnosis. Increased FDG uptake in the cervical and supraclavicular BAT regions led to diagnostic confusion, mimicking diffuse regional metastasis. Careful interpretation of PET/CT imaging, with fusion of anatomical and functional data, was essential to differentiate hypermetabolic BAT from malignant disease. Increased BAT FDG uptake, particularly in patients using GLP-1 receptor agonists, can complicate the evaluation of head and neck cancer. Awareness of this interaction is critical to avoid misdiagnosis and overtreatment. Laryngoscope, 135:741-743, 2025.
Collapse
Affiliation(s)
- Daron B Harrison
- Department of Otolaryngology-Head & Neck Surgery, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - Alisa L Phillips
- Department of Otolaryngology-Head & Neck Surgery, Oregon Health & Science University, Portland, Tennessee, U.S.A
| | - James B Tansey
- Department of Otolaryngology-Head & Neck Surgery, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - Travis J Clarke
- Department of Otolaryngology-Head & Neck Surgery, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - C B Wood
- Department of Otolaryngology-Head & Neck Surgery, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - Lucien Nedzi
- Department of Radiation Oncology, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - David L Schwartz
- Department of Radiation Oncology, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - Liza Makowski
- Department of Hematology Oncology, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
- Center for Cancer Research, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - D N Hayes
- Department of Hematology Oncology, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
- Center for Cancer Research, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| | - Grace Newman
- Department of Nuclear Medicine, Methodist Le Bonheur Healthcare, Memphis Radiological PC, Memphis, Tennessee, U.S.A
| | - John P Gleysteen
- Department of Otolaryngology-Head & Neck Surgery, University of Tennessee Health Science Center College of Medicine, Memphis, Tennessee, U.S.A
| |
Collapse
|
44
|
Zhang Y, Zhou S, Zhao R, Huang Y, Wang Y. Chronic cold exposure reprograms feeding-regulated LPL activity in white adipose tissues through hepatic ANGPTL3 and ANGPTL8. LIFE METABOLISM 2025; 4:loae037. [PMID: 39872988 PMCID: PMC11770819 DOI: 10.1093/lifemeta/loae037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 01/30/2025]
Abstract
Graphical Abstract Lipoprotein lipase (LPL) mediates peripheral tissue triglyceride (TG) uptake. Hepatic ANGPTL3 (A3) and ANGPTL8 (A8) form a complex and inhibit LPL activity in the white adipose tissue (WAT) via systematic circulation. ANGPTL4 (A4) is expressed in WAT and inhibits LPL activity locally. Feeding increases hepatic A8 expression and increases its inhibition for WAT LPL activity together with A3, while feeding suppresses WAT A4 expression and releases its inhibition on LPL. At room temperature, the feeding-suppressed A4 overrides the feeding-increased A3/A8, resulting in increased LPL activity in WAT by food intake. Browning improves hepatic insulin sensitivity and increases postprandial A8 expression. The feeding-increased A3/A8 overrides the feeding-suppressed A4, resulting in suppressed LPL activity in WAT by food intake. This reprogrammed LPL regulation plays an important role in reprogramming TG metabolism during adipose tissue browning.
Collapse
Affiliation(s)
- Yiliang Zhang
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Shengyang Zhou
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Runming Zhao
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yingzhen Huang
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, Department of Biochemistry, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, Hubei 430072, China
| |
Collapse
|
45
|
Unlu Y, Piaggi P, Stinson EJ, De Baca TC, Rodzevik TL, Walter M, Fry H, Krakoff J, Chang DC. Cold induces increased ad libitum energy intake independent of changes in energy expenditure: a controlled crossover trial in adults. Am J Clin Nutr 2025; 121:293-303. [PMID: 39675563 PMCID: PMC11863325 DOI: 10.1016/j.ajcnut.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/20/2024] [Accepted: 12/11/2024] [Indexed: 12/17/2024] Open
Abstract
BACKGROUND Measures of energy metabolism (energy expenditure [EE], respiratory exchange ratio [RER]) have been associated with ad libitum energy intake (EI) and weight gain in previous observational studies, suggesting that energy-sensing mechanisms drive EI to meet metabolic energy demands. OBJECTIVES We aimed to employ mild cold exposure as an intervention to alter energy metabolism and evaluate its causal effects on concurrent and next day ad libitum EI. METHODS In a controlled crossover study, 47 volunteers (16 female; age 37.2 ± 10.7 y; body mass index 32.4 ± 8.6 kg/m2) completed four 24-h EE measurements in a respiratory chamber. Participants ate ad libitum for 24-h using an objective vending machine paradigm while in the chamber during both cold exposure (19°C) and thermoneutral conditions (23.5°C) and after 2 eucaloric chamber stays with exposure to each temperature. Energy metabolism changes were calculated from eucaloric conditions (cold compared with thermoneutral). RESULTS Compared with thermoneutral conditions, participants consumed 13% more while residing in the chamber during cold (mean difference: 411 ± 987 kcal/d, P = 0.006), but not the day after cold exposure. Neither eucaloric EE, RER, nor carbohydrate oxidation (CARBOX) was significantly changed by cold exposure. However, greater increases in RER and CARBOX during cold exposure were associated with greater ad libitum EI on the day after cold exposure (r = 0.29, P = 0.049 and r = 0.33, P = 0.02), but not with EI during cold exposure. Cold-induced changes in 24-h EE were not associated with changes in ad libitum EI during or after cold exposure. CONCLUSION Ad libitum EI increased during but not after mild cold exposure. There was an effect of 24-h RER and CARBOX during cold exposure that was related to greater ad libitum EI after cold. These results indicate an acute effect of cold on concurrent EI independent of changes in metabolic rate, but also a residual influence of cold on subsequent EI via fuel preference. This trial was registered at clinicaltrials.gov as NCT02939404.
Collapse
Affiliation(s)
- Yigit Unlu
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Paolo Piaggi
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States; Department of Information Engineering, University of Pisa, Pisa, Italy.
| | - Emma J Stinson
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Tomás Cabeza De Baca
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Theresa L Rodzevik
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Mary Walter
- Clinical Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Hannah Fry
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Jonathan Krakoff
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| | - Douglas C Chang
- Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Phoenix, AZ, United States
| |
Collapse
|
46
|
Takada M, Kawarasaki S, Kwon J, Ni Z, Takahashi H, Inoue K, Goto T. Lipid metabolism and food ingredients from the perspective of thermogenic adipocytes. Biosci Biotechnol Biochem 2025; 89:193-200. [PMID: 39521946 DOI: 10.1093/bbb/zbae162] [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: 09/23/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
The high heat-producing capacity of brown and beige adipocytes, collectively known as thermogenic adipocytes, contributes to whole-body energy expenditure and is an attractive target for the management of obesity. It has been revealed that the functions of thermogenic adipocytes are important for the regulation of whole-body carbohydrate and lipid metabolism, and the activation of thermogenic adipocytes seems to have beneficial effects for the management of obesity-related metabolic disorders, such as dyslipidemia. Recent studies have showed that specific food ingredients have the potential to activate thermogenic adipocytes via various mechanisms. Some of these are effective not only in rodents, but also in humans, and effective prevention of obesity using these food ingredients is expected. In this review, we introduce the recent findings on the regulatory mechanisms of lipid metabolism by thermogenic adipocytes and food ingredients, demonstrating the potential to activate thermogenic adipocytes and their underlying mechanisms.
Collapse
Affiliation(s)
- Mai Takada
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Satoko Kawarasaki
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Jungin Kwon
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Zheng Ni
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Haruya Takahashi
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
| | - Kazuo Inoue
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| | - Tsuyoshi Goto
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Uji, Kyoto, Japan
- Research Unit for Physiological Chemistry, The Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, Japan
| |
Collapse
|
47
|
Zhang M, Traspov A, Yang J, Zheng M, Kharzinova VR, Ai H, Zinovieva NA, Huang L. Genomic and transcriptomic insights into vitamin A-induced thermogenesis and gene reuse as a cold adaptation strategy in wild boars. Commun Biol 2025; 8:116. [PMID: 39856249 PMCID: PMC11759952 DOI: 10.1038/s42003-025-07536-8] [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: 09/08/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Wild boars inhabit diverse climates, including frigid regions like Siberia, but their migration history and cold adaptation mechanisms into high latitudes remain poorly understood. We constructed the most comprehensive wild boar whole-genome variant dataset to date, comprising 124 samples from tropical to frigid zones, among which 47 Russian, 8 South Chinese and 3 Vietnamese wild boars were newly supplemented. We also gathered 75 high-quality RNA-seq datasets from 10 tissues of 6 wild boars from Russia and 6 from southern China. Demographic analysis revealed the appearance of Russian wild boars in Far East of Asia (RUA) and Europe (RUE) after the last glacial maximum till ~ 10 thousand years ago. Recent gene flow (<100 years) from RUA to RUE reflects human-mediated introductions. Cold-region wild boars exhibit strong selection signatures indicative of genetic adaptation to cold climates. Further pathway and transcriptomic analyses reveal a novel cold resistance mechanism centered on enhanced vitamin A metabolism and catalysis, involving the reuse of UGT2B31 and rhythm regulation by ANGPTL8, RLN3 and ZBTB20. This may compensate for the pig's lack of brown fat/UCP1 thermogenesis. These findings provide new insights into the molecular basis of cold adaptation and improve our understanding of Eurasian wild boar migration history.
Collapse
Affiliation(s)
- Mingpeng Zhang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
- Key Laboratory of Biodiversity Conservation and Bioresource Utilization of Jiangxi Province, College of Life Sciences, Jiangxi Normal University, Nanchang, Jiangxi Province, P.R. China
| | - Aleksei Traspov
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Jiawen Yang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Min Zheng
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China
| | - Veronika R Kharzinova
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia
| | - Huashui Ai
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| | - Natalia A Zinovieva
- L.K. Ernst Federal Research Center for Animal Husbandry, Dubrovitsy, Podolsk Municipal District, Moscow Region, Podolsk, Russia.
| | - Lusheng Huang
- National Key Laboratory for Swine genetic improvement and production technology, Ministry of Science and Technology of China, Jiangxi Agricultural University, Nanchang, Jiangxi Province, P.R. China.
| |
Collapse
|
48
|
Pan L, Ai S, Shi X, Tong X, Spanos M, Li G, Cretoiu D, Gao J, Zhou Q, Xiao J. ExerGeneDB: A physical exercise-regulated differential gene expression database. JOURNAL OF SPORT AND HEALTH SCIENCE 2025; 14:101027. [PMID: 39827990 PMCID: PMC12013164 DOI: 10.1016/j.jshs.2025.101027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/10/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Exercise induces molecular changes that involve multiple organs and tissues. Moreover, these changes are modulated by various exercise parameters-such as intensity, frequency, mode, and duration-as well as by clinical features like gender, age, and body mass index (BMI), each eliciting distinct biological effects. To assist exercise researchers in understanding these changes from a comprehensive perspective that includes multiple organs, diverse exercise regimens, and a range of clinical features, we developed Exercise Regulated Genes Database (ExerGeneDB), a database of exercise-regulated differential genes. METHODS ExerGeneDB aggregated publicly available exercise-related sequencing datasets and subjected them to uniform quality control and preprocessing. The data, encompassing a variety of types, were organized into a specialized database of exercise-regulated genes. Notably, ExerGeneDB conducted differential analyses on this collected data, leveraging curated clinical information and accounting for important factors such as gender, age, and BMI. RESULTS ExerGeneDB has assembled 1692 samples from rats and mice as well as 4492 human samples. It contains data from various tissues and organs, such as skeletal muscle, blood, adipose tissue, intestine, heart, liver, spleen, lungs, kidneys, brain, spinal cord, bone marrow, and bones. ExerGeneDB features bulk ribonucleic acid sequencing (RNA-seq) (including non-coding RNA (ncRNA) and protein-coding RNA), microarray (including ncRNA and protein-coding RNA), and single cell RNA-seq data. CONCLUSION ExerGeneDB compiles and re-analyzes exercise-related data with a focus on clinical information. This has culminated in the creation of an interactive database for exercise regulation genes. The website for ExerGeneDB can be found at: https://exergenedb.com.
Collapse
Affiliation(s)
- Ling Pan
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Songwei Ai
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Xiaohui Shi
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Xiaolan Tong
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Michail Spanos
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Guoping Li
- Cardiovascular Division of the Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Dragos Cretoiu
- Department of Medical Genetics, Carol Davila University of Medicine and Pharmacy, Bucharest 020031, Romania; Materno-Fetal Assistance Excellence Unit, Alessandrescu-Rusescu National Institute for Mother and Child Health, Bucharest 011062, Romania
| | - Juan Gao
- Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China
| | - Qiulian Zhou
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.
| | - Junjie Xiao
- Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, Joint International Research Laboratory of Biomaterials and Biotechnology in Organ Repair (Ministry of Education), School of Life Science, Shanghai University, Shanghai 200444, China; Cardiac Regeneration and Ageing Lab, Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.
| |
Collapse
|
49
|
van Eenige R, Hoekx CA, Sardjoe Mishre ASD, Straat ME, Boon MR, Martinez‐Tellez B, Rensen PCN, Kan HE. Cold exposure and thermoneutrality similarly reduce supraclavicular brown adipose tissue fat fraction in fasted young lean adults. FASEB J 2025; 39:e70307. [PMID: 39797666 PMCID: PMC11724390 DOI: 10.1096/fj.202402415r] [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: 10/10/2024] [Revised: 12/05/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025]
Abstract
Brown adipose tissue (BAT) is a metabolically highly active tissue that dissipates energy stored within its intracellular triglyceride droplets as heat. Others have previously utilized MRI to show that the fat fraction of human supraclavicular BAT (scBAT) decreases upon cold exposure, compared with baseline (i.e., pre-cooling). However, comparisons to a control group that was not exposed to cold are largely lacking. We recently developed a non-invasive dynamic MRI protocol that allows for quantifying scBAT fat fraction changes over time. Here, we aimed to study the effect of cold exposure versus thermoneutrality on fat fraction changes in human scBAT. Ten young (mean age: 21.5 ± 0.7 years), lean (mean BMI: 21.7 ± 0.5 kg/m2), 12 h-fasted volunteers (9 females; 1 male) underwent up to 70 consecutive MRI scans each on two separate study visits in a cross-over design. Participants were exposed to a temperature of 32°C for 10 scans (i.e., ±16 min), which was then either lowered to 18°C (i.e., cold exposure) or was maintained at 32°C (i.e., thermoneutrality). Dynamic fat fraction changes were quantified, and self-reported thermal perception scores were monitored. The fat fraction in scBAT decreased over time upon cold exposure (r = -.222, p < .001). Interestingly however, we also observed a decrease in scBAT fat fraction over time upon thermoneutrality (r = -.212, p < .001). No difference was observed between the two temperature conditions (p = .55), while self-reported thermal perception scores were consistently higher (i.e., colder) upon cold exposure. In the trapezius muscle and the humerus bone as control tissues, the fat fraction was unaffected in both temperature conditions. The fat fraction in subcutaneous white adipose tissue (sWAT) however, also decreased over time upon cold exposure (r = -.270, p < .001) and during thermoneutrality (r = -.190, p < .001), again with no difference (p = .92) between the two temperature conditions. In conclusion, our results show that in 12 h-fasted, healthy individuals cold exposure and thermoneutrality similarly reduce the fat fraction within scBAT and sWAT. While we interpret that the cold exposure was sufficient to induce thermogenesis, we suggest that an increased lipolytic activity within adipocytes, as a consequence of fasting, may be the primary cause of the decreased fat fraction in both sWAT and scBAT in our study. The current study highlights the potential influence of fasting on the fat fraction in scBAT and stresses the importance of inclusion of a thermoneutral control group in studies investigating the BAT-modulating effect of cold exposure.
Collapse
Affiliation(s)
- Robin van Eenige
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
| | - Carlijn A. Hoekx
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | | | - Maaike E. Straat
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Mariëtte R. Boon
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Borja Martinez‐Tellez
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
- Department of Nursing Physiotherapy and Medicine, SPORT Research Group (CTS‐1024), CIBIS Research CenterUniversity of AlmeríaAlmeríaSpain
- Biomedical Research UnitTorrecárdenas University HospitalAlmeríaSpain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN)Instituto de Salud Carlos IIIGranadaSpain
| | - Patrick C. N. Rensen
- Division of Endocrinology, Department of MedicineLeiden University Medical CenterLeidenThe Netherlands
- Einthoven Laboratory for Experimental Vascular MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Hermien E. Kan
- Department of Radiology, C.J. Gorter MRI CenterLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
50
|
Cypess AM, Cannon B, Nedergaard J, Kazak L, Chang DC, Krakoff J, Tseng YH, Schéele C, Boucher J, Petrovic N, Blondin DP, Carpentier AC, Virtanen KA, Kooijman S, Rensen PCN, Cero C, Kajimura S. Emerging debates and resolutions in brown adipose tissue research. Cell Metab 2025; 37:12-33. [PMID: 39644896 PMCID: PMC11710994 DOI: 10.1016/j.cmet.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/28/2024] [Accepted: 11/01/2024] [Indexed: 12/09/2024]
Abstract
Until two decades ago, brown adipose tissue (BAT) was studied primarily as a thermogenic organ of small rodents in the context of cold adaptation. The discovery of functional human BAT has opened new opportunities to understand its physiological role in energy balance and therapeutic applications for metabolic disorders. Significantly, the role of BAT extends far beyond thermogenesis, including glucose and lipid homeostasis, by releasing mediators that communicate with other cells and organs. The field has made major advances by using new model systems, ranging from subcellular studies to clinical trials, which have also led to debates. In this perspective, we identify six fundamental issues that are currently controversial and comprise dichotomous models. Each side presents supporting evidence and, critically, the necessary methods and falsifiable experiments that would resolve the dispute. With this collaborative approach, the field will continue to productively advance the understanding of BAT physiology, appreciate the importance of thermogenic adipocytes as a central area of ongoing research, and realize the therapeutic potential.
Collapse
Affiliation(s)
- Aaron M Cypess
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Barbara Cannon
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Jan Nedergaard
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Lawrence Kazak
- Rosalind & Morris Goodman Cancer Institute, McGill University, Montreal, QC H3A 1A3, Canada; Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Douglas C Chang
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Jonathan Krakoff
- Phoenix Epidemiology and Clinical Research Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Phoenix, AZ 85016, USA
| | - Yu-Hua Tseng
- Joslin Diabetes Center, Harvard Medical School, Boston, MA 02115, USA
| | - Camilla Schéele
- Novo Nordisk Foundation Center for Basic Metabolic Research, The Center of Inflammation and Metabolism and the Center for Physical Activity Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Natasa Petrovic
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, 106 91 Stockholm, Sweden
| | - Denis P Blondin
- Division of Neurology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André C Carpentier
- Division of Endocrinology, Department of Medicine, Centre de recherche du Centre hospitalier universitaire de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, the Netherlands
| | - Cheryl Cero
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Howard Hughes Medical Institute, Boston, MA, USA.
| |
Collapse
|